WO2014145729A2 - Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same - Google Patents

Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same Download PDF

Info

Publication number
WO2014145729A2
WO2014145729A2 PCT/US2014/030538 US2014030538W WO2014145729A2 WO 2014145729 A2 WO2014145729 A2 WO 2014145729A2 US 2014030538 W US2014030538 W US 2014030538W WO 2014145729 A2 WO2014145729 A2 WO 2014145729A2
Authority
WO
WIPO (PCT)
Prior art keywords
tissue
dkp
stem cells
cells
cell
Prior art date
Application number
PCT/US2014/030538
Other languages
English (en)
French (fr)
Other versions
WO2014145729A3 (en
Inventor
David Bar-Or
Greg Thomas
Original Assignee
Ampio Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to JP2016503416A priority Critical patent/JP6588005B2/ja
Priority to SG11201506267XA priority patent/SG11201506267XA/en
Priority to KR1020157029761A priority patent/KR20150132508A/ko
Priority to BR112015020469A priority patent/BR112015020469A2/pt
Priority to EP14763038.8A priority patent/EP2968315B1/en
Priority to AU2014232728A priority patent/AU2014232728B2/en
Priority to MX2015010937A priority patent/MX2015010937A/es
Priority to EA201500943A priority patent/EA201500943A1/ru
Application filed by Ampio Pharmaceuticals, Inc. filed Critical Ampio Pharmaceuticals, Inc.
Priority to CN201480015832.9A priority patent/CN105101965B/zh
Priority to CA2906864A priority patent/CA2906864A1/en
Priority to NZ712630A priority patent/NZ712630A/en
Publication of WO2014145729A2 publication Critical patent/WO2014145729A2/en
Publication of WO2014145729A3 publication Critical patent/WO2014145729A3/en
Priority to IL240473A priority patent/IL240473A0/en
Priority to PH12015502048A priority patent/PH12015502048A1/en
Priority to ZA2015/07291A priority patent/ZA201507291B/en
Priority to HK16102085.4A priority patent/HK1214143A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/32Bones; Osteocytes; Osteoblasts; Tendons; Tenocytes; Teeth; Odontoblasts; Cartilage; Chondrocytes; Synovial membrane
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/999Small molecules not provided for elsewhere

Definitions

  • This invention relates to the field of stem cell technology. More particularly, the invention describes compositions for the mobilization, homing, expansion and/or differentiation of stem cells and methods of using the same.
  • Stem cells have the ability to divide for indefinite periods in culture and to become a wide variety of specialized cell and tissue types, which can then be used for basic research, drug discovery, and treatment (or prevention) of many diseases. Stem cells are typically divided into two main groups: adult stem cells and embryonic stem cells.
  • Adult stem cells are undifferentiated but are present in differentiated tissues, and are capable of differentiation into the cell types from which tissue the adult stem cell originated.
  • Adult stem cells have been derived from various sources, such as the nervous system, bone marrow; adipose tissue, dermis, pancreas and liver. Stem cells have also been isolated from umbilical cord and placenta.
  • stem cells of the adult type are also found in smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone spongy tissue, cartilage tissue, pancreatic ductal tissue, spleen tissue, thymus tissue, tonsil tissue, Peyer's patch tissue, lymph nodes tissue, thyroid tissue, epidermis tissue, dermis tissue, subcutaneous tissue, heart tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophagus tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterus tissue, eye tissue (including retinal tissue), lung tissue, testicular tissue, ovarian tissue, prostate tissue, connective tissue, endocrine tissue, and mesentery tissue.
  • Embryonic stem cells are undifferentiated cells derived from the embryo. Typically, these cells are extracted from the inner cell mass of a blastocyte and when cultured under the unique conditions, either alone or in combination with a variety of feeder cells, the embryonic stem cells maintain euploid karyotype, do not undergo senescence, and retain the ability to differentiate into cells of the endodermal, ectodermal, and mesodermal lineages.
  • MSCs Mesenchymal stem cells
  • MSCs Mesenchymal stem cells
  • the major source of MSCs is isolation from bone marrow and other tissues, such as adipose tissue. Due to their scarcity in adult tissues, MSCs are normally isolated only in small numbers, however, they have an extensive capacity for proliferation and can be readily expanded in culture to generate clinically-relevant numbers of cells through multiple passages. These cells have been shown to support tissue repair and regeneration, making them a promising tool for various therapeutic applications.
  • stem cells One of the earliest clinical uses of stem cells was for performing bone marrow transplants in patients with hematological malignancies in which hematopoietic stem cells derived from the donor bone marrow were administered into the recipient subsequent to treating the recipient with a dose of radiation and/or chemotherapy in order to ablate not only the hematological malignancy but also non-malignant hematopoiesis.
  • the administration of non-malignant hematopoietic stem cells results in donor-specific hematopoiesis, and in some patients, cure of the malignancy. This was first described in 1957 in patients with acute leukemia following myeloablation.
  • Stem cells have also been utilized as an autologous bone marrow transplant in patients administered high doses of chemotherapy and/or radiation therapy for the treatment of solid tumors, in order to restore bone marrow.
  • the use of autologous hematopoietic cell transplants combined with high dose chemo/radiotherapy for solid tumors has been extensively investigated for breast, colon, lung, nasopharyngeal cancer, and other types of cancers.
  • autologous stem cells have also been performed for a variety of autoimmune indications, including rheumatoid arthritis, multiple sclerosis, systemic lupus erythromatosis, and systemic sclerosis.
  • stem cell culture systems used in these treatments e.g., liquid static culture, semi-solid culture and long term bone marrow culture
  • animal sera e.g., fetal bovine serum or horse serum
  • serum in the culture of hematopoietic cells is disadvantageous for several reasons. Serum is a major source of undefined differentiation factors and thus tends to promote hematopoietic cell differentiation, rather than expansion. The efficiency of serum varies between lots of serum. Some lots of serum have been found to be toxic to cells. Moreover, serum may be contaminated with infectious agents such as mycoplasma, bacteriophage, and viruses. These problems cause inconsistencies in the growth- supporting properties of the medium, making standardization of stem cell production processes difficult and making the interpretation of experiments carried out in serum- containing media difficult. Thus, the use of serum represents a major obstacle for the clinical implementation of stem cell-related therapies.
  • defined serum-free media and methods for (i) isolating stem cells and (ii) expanding these cells for an extended period of time through multiple passages while maintaining their multi-lineage differentiation potential.
  • the creation of defined serum- free media and methods should provide a robust platform that will help to enable the clinical implementation of stem cell-based therapies.
  • One embodiment of the present invention is a method to cause an effect selected from the group consisting of stem cell mobilization, stem cell homing, stem cell expansion, and stem cell differentiation in a subject.
  • the method includes administering DA-DKP to a subject in need thereof.
  • the DA-DKP can be administered in various formulations, be administered by various routes of administration and be produced by various methods.
  • the DA-DKP can be administered as a pharmaceutical composition that includes DA-DKP.
  • Such pharmaceutical compositions can also include N-acetyl tryptophan, caprylate and/or caprylic acid.
  • Such pharmaceutical compositions can include a fraction of human serum albumin, wherein substantially all of the albumin has been removed from the fraction or can include a low molecular weight fraction of human serum albumin, such as a less than 5000 molecular weight fraction.
  • Such fractions of human serum albumin can be produced by filtration.
  • the pharmaceutical composition can be prepared by heating human serum albumin under conditions effective to cause the formation of DA-DKP or by a process including contacting human serum albumin with an enzyme that cleaves the two N-terminal amino acids of human albumin under conditions effective to produce DA-DKP.
  • the DA-DKP can be a synthetic DA-DKP.
  • the DA-DKP can be administered locally to the subject.
  • the site of local administration can be selected from a joint, a surgical site, a site of a segmented skeletal gap or non-union fracture, a wound, an ulcer, and an inflammatory skin rash.
  • the DA-DKP can be administered as a parenteral formulation, can be administered systemically to the subject or can be administered as an oral dosage formulation.
  • the DA-DKP can be formulated as part of, or on an implantable device, such as one selected from a sponge, biocompatible polymer, bioerodible polymer, putty, gel, bone matrix, artificial bone matrix, bolt, screw, endotracheal tube, stent, contact lense, pacemaker, central IV tube, foley catheter, and intracranial device.
  • an implantable device such as one selected from a sponge, biocompatible polymer, bioerodible polymer, putty, gel, bone matrix, artificial bone matrix, bolt, screw, endotracheal tube, stent, contact lense, pacemaker, central IV tube, foley catheter, and intracranial device.
  • administration of DA-DKP can increase production of CXCR4, decrease production of CXCL12, increase production of MMP14 or MMP13, increase production of aggrecan, increase production of SDF1, increase production of collagen 2A1 or any combination of the foregoing.
  • administration of DA-DKP can decrease production of a protein selected from the group consisting of MAPK-activated protein kinase 3, beta-adrenergic receptor kinase 1, ADAM metallopeptidase with thrombodpondin type I motif, MAPK-activated protein kinase 2, C- Src kinase, Macrophage Scavenger Receptor, Noggin, Tyrosine kinase Bruton, Glycogen synthase kinase-3 alpha/beta, Glycogen synthase kinase-3 alpha/beta, HSP 90 alpha/beta, HSP 90 alpha/beta, Phosphoinositide-3 -kinase, catalytic subunit alpha, and Eukaryotic translation initiation factor 4A, Fibroblast Growth Factor 17 and combinations thereof.
  • MAPK-activated protein kinase 3 beta-adrenergic receptor kinas
  • administration of DA-DKP can decrease production of a protein selected from MAPK-activated protein kinase 3, Noggin, Phosphoinositide-3 -kinase, catalytic subunit alpha, and combinations thereof.
  • Administration of DA-DKP can also increase production of a protein selected from Clusterin (Apo lipoprotein J), Prothrombin, C1QBP (Hyaluronan binding protein 1), TNFSF 15 (VEGF inhibitor), Mammaglobin 2, MIP3b (CCL 19), MCP 1 (CCL 2), PTHrP, Spondin 1, Elafm (elastase inhibitor), IL 11, NPS-PLA2, CFC 1 (cryptic protein), Testican 1 (SPOCK 1), Angiogenin, URB, MMP-3, IP 10 (cxcl 10), BSSP 4, IL 8 (cxcl 8), RSP02, Cystatin C, bFGF, Factor H, Coagulation Factor IX, S
  • Administration of DA-DKP can also increase production of a protein selected from Clusterin (Apolipoprotein J), C1QBP (Hyaluronan binding protein 1), MCP 1 (CCL 2), PTHrP, Elafm (elastase inhibitor), IL 11, MMP-3, bFGF, SAA, TIMP-1, Semaphorin 3 A, and combinations thereof.
  • Clusterin Apolipoprotein J
  • C1QBP Hydrophilin 1
  • MCP 1 CCL 2
  • PTHrP Elafm (elastase inhibitor)
  • IL 11 MMP-3
  • SAA TIMP-1
  • Semaphorin 3 A Semaphorin 3 A
  • Administration of DA-DKP can also decrease production of a protein selected from MAPK-activated protein kinase 3, Noggin, Phosphoinositide-3 - kinase, catalytic subunit alpha, and combinations thereof and increase production of a protein selected from the group consisting of Clusterin (Apolipoprotein J), C1QBP (Hyaluronan binding protein 1), MCP 1 (CCL 2), PTHrP, Elafm (elastase inhibitor), IL 11, MMP-3, bFGF, SAA, TIMP-1, Semaphorin 3 A, and combinations thereof. Further, administration of DA-DKP can down regulates Akt pathways in the subject.
  • a protein selected from MAPK-activated protein kinase 3, Noggin, Phosphoinositide-3 - kinase, catalytic subunit alpha, and combinations thereof and increase production of a protein selected from the group consisting of Clusterin (Apolipoprotein J), C1Q
  • Another embodiment of the present invention is a method of stimulating chondrogenesis in a subject by administering a pharmaceutical composition that includes DA-DKP and optionally, a component selected from N-acetyl tryptophan, caprylate, caprylic acid, and combinations thereof to a subject in need thereof.
  • chondrogenesis can be stimulated in a stem cell, such as a stem cell selected from a progenitor cell and mesenchymal stem call (MSC).
  • the stimulation of chondrogenesis can promote cartilage, bone, and/or ligament repair or induce repair or regeneration of chondral tissue, in the subject.
  • the chondrogenesis can treat or ameliorate a chondrogenic disease in the subject, and the chondrogenic disease can be a congenital cartilage disease, degenerative or fibrotic joint, rheumatoid arthritis or osteoarthritis.
  • the chondrogenesis can treat or repair a condition selected from a cartilage defect, a skeletal defect, or a fracture arising from trauma or surgery.
  • the DA-DKP administration can stimulate the formation of new bone or cartilage tissue.
  • the administration can include stimulating stem cells ex vivo and then administering the stimulated stem cells to the subject.
  • the stem cells can be stimulated ex vivo by culturing a population of stem cells of chondrocyte lineage with DA-DKP, or composition comprising a DA-DKP, for a time sufficient to stimulate chondrogenesis, and the step of administering includes implanting the stimulated cells into a desired site in the subject.
  • the chondrogenesis can result in increased production of collagen, type 2A1 collagen, type 1A1 collagen, or a 2-fold, 4-fold, 10-fold, 20-fold, or 25 -fold increase in the production of collagen.
  • Further embodiments of the present invention include use of DA-DKP or a composition including DA-DKP in the preparation of a medicament for the stimulation of chondrogenesis in a mammal, and use of DA-DKP or a composition including DA-DKP for the stimulation of chondrogenesis in a mammal.
  • Another embodiment of the invention is a method of stimulating development of nervous tissue in a subject by administering a pharmaceutical composition that includes DA-DKP and optionally, a component selected from N-acetyl tryptophan, caprylate, caprylic acid, and combinations thereof to a subject in need thereof.
  • the development of nervous tissue can be stimulated in a stem cell, such as aprogenitor cell or mesenchymal stem call (MSC).
  • MSC mesenchymal stem call
  • the stimulation of development of nervous tissue can promote brain, spinal cord, and/or peripheral nerve repair or induce repair or regeneration of neurons, neuroglia, and/or astrocytes in the subject.
  • the development of nervous tissue can treat or ameliorate a disease of the central nervous system and/or a disease of the peripheral nervous system in the subject, such as a neurodegenerative disease. Also, the development of nervous tissue can treat or repair a condition selected from an injury arising from trauma or surgery.
  • the administration can include stimulating stem cells ex vivo and then administering the stimulated stem cells to the subject.
  • the stem cells can be stimulated ex vivo by culturing a population of stem cells of neuron, neuroglia, and/or astrocyte lineage with DA-DKP, or composition that includes DA-DKP, for a time sufficient to stimulate development of nervous tissue, and the step of administering includes implanting the stimulated cells into a desired site in the subject.
  • Further embodiments of the present invention include the use of DA-DKP or a composition that includes DA-DKP in the preparation of a medicament for the stimulation of development of nervous tissue in a subject, and use of DA-DKP or a composition that includes DA-DKP for the stimulation of development of nervous tissue in a subject.
  • a further embodiment of the invention is a method of stimulating development of tissue in a subject by administering DA-DKP to a subject in need thereof.
  • the tissue can be selected from nervous system tissue, adipose tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone spongy tissue, cartilage tissue, pancreatic ductal tissue, spleen tissue, thymus tissue, tonsil tissue, Peyer's patch tissue, lymph nodes tissue, thyroid tissue, epidermis tissue, dermis tissue, subcutaneous tissue, heart tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophagus tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterus tissue, eye tissue, lung tissue, testicular tissue, ovarian tissue, prostate tissue, connective tissue, endocrine tissue, and mesentery tissue.
  • FIG. 1 For embodiments of the invention, are a serum-free, eukaryotic cell culture media supplement that includes DA-DKP, wherein a cell culture medium supplemented with said supplement is capable of supporting the expansion of stem cells, and a composition that includes stem cells in a serum-free media supplemented with the media supplement.
  • Another embodiment of the invention is a method of expanding stem cells, that includes contacting the stem cells with DA-DKP, and culturing the stem cells under conditions suitable to facilitate the expansion of the stem cells.
  • a further embodiment of the invention is method of providing stem cells to a mammal, by contacting stem cells with DA-DKP, cultivating the stem cells under conditions suitable to facilitate the expansion of the cells; and introducing the expanded cells into a subject.
  • a still further embodiment of the invention is a method of causing stem cells to differentiate into a particular type of cell by contacting stem cells with DA-DKP, cultivating the stem cells under conditions suitable to facilitate the expansion of the stem cells, and adding one or more differentiation factors or changing culturing conditions to induce differentiation of the stem cells to form a different type of cell.
  • Another embodiment of the invention is a method of providing differentiated stem cells, to a subject.
  • the method includes contacting stem cells with DA-DKP, cultivating the stem cells under conditions suitable to facilitate the expansion of the stem cells, and adding one or more differentiation factors or changing culturing conditions to induce differentiation of cells to form a different type of cell.
  • the method further includes introducing the differentiated cells into the subject.
  • Figure 1 illustrates the mean change in pain scores for osteoarthritis patients receiving AmpionTM compared to patients receiving saline.
  • Figure 2 are photographs of cell aggregates from stem cells treated with saline, dexamethasone, DA-DKP and AmpionTM.
  • Figure 3 illustrates the enhancement of collagen and aggrecan transcription by stem cells treated with DA-DKP as compared to saline.
  • Figure 4 illustrates the increase in production of CXCR4 by mesenchymal stem cells grown in 3D culture by AmpionTM compared to saline.
  • Figure 5 illustrates the decrease in production of CXCL12 by mesenchymal stem cells grown in 3D culture by AmpionTM compared to saline.
  • Figure 6 illustrates the effect of AmpionTM on the migration of stem cells in vitro.
  • Figure 7 demonstrates the effect of AmpionTM on the transcription of Collagen 2A1 by bone marrow derived human mesenchymal stem cells.
  • Figure 8 demonstrates the effect of Ampion on the transcription of Aggrecan by bone marrow derived human mesenchymal stem cells.
  • Figure 9 demonstrates the effect of AmpionTM on the transcription of GAPDH by bone marrow derived human mesenchymal stem cells.
  • Figure 10 illustrates the patient disposition of the clinical trial described in
  • Figure 11 illustrates the reduction in pain for osteoarthritis patients receiving injections of LMWF-5A as compared to vehicle control.
  • the present invention is drawn to methods for causing stem cell mobilization, homing, and/or differentiation in a subject in need thereof.
  • the present invention is further directed to compositions that safely and effectively promote expansion of stem cells and methods of using such compositions and expanded cells in the treatment of disease states amenable to treatment with stem cells.
  • albumin substitute refers to any compound which may be used in place of human serum albumin in the compositions of the invention to give substantially similar or better results as human serum albumin.
  • the albumin is of human origin.
  • the albumin is human serum albumin.
  • expand or “expansion” refers to the growth and division, and not the differentiation, of stem cells in culture.
  • differentiation refers to the development of a cell of a particular type into a cell of another type.
  • the development of a pluripotent hematopoietic stem cell into a myeloid precursor is an example of differentiation.
  • the development of precursor cell into another type of cell is an example of differentiation.
  • stem cell generally refers to any cells that have the ability to divide for indefinite periods of time and to give rise to specialized cells.
  • totipotent cells such as an embryonic stem cell, an extra-embryonic stem cell, a cloned stem cell, a parthenogenesis derived cell, a cell reprogrammed to possess totipotent properties, or a primordial germ cell;
  • pluripotent cell such as a hematopoietic stem cell, an adipose derived stem cell, a mesenchymal stem cell, a cord blood stem cell, a placentally derived stem cell, an exfoliated tooth derived stem cells, a hair follicle stem cell or a neural stem cell; and c) a tissue specific progenitor cell such as a precursor cell for the neuronal, hepatic, nephrogenic, adipogenic, osteoblastic, osteoclastic, alve
  • the cells can be derived, for example, from tissues such as pancreatic tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone marrow tissue, bone spongy tissue, cartilage tissue, liver tissue, pancreas tissue, pancreatic ductal tissue, spleen tissue, thymus tissue, Peyer's patch tissue, lymph nodes tissue, thyroid tissue, epidermis tissue, dermis tissue, subcutaneous tissue, heart tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophagus tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterus tissue, eye tissue (including retinal tissue), lung tissue, testicular tissue, ovarian tissue, prostate tissue, connective tissue, endocrine tissue, and mesentery tissue. Specific stem cells are described below in the section entitled Ex Vivo Treatment Methods.
  • totipotent cells refers to mammalian cells that have the potential to become any cell type in the adult body and any cell type of the extraembryonic membranes (e.g., placenta). Totipotent cells are the fertilized egg and approximately the first 4 cells produced by its cleavage.
  • pluripotent stem cells refers to true stem cells with the potential to make any differentiated cell in the body, but cannot contribute to making the components of the extraembryonic membranes which are derived from the trophoblast. The amnion develops from the epiblast, not the trophoblast.
  • Three types of pluripotent stem cells have been confirmed: Embryonic Stem (ES) Cells (may also be totipotent in primates), Embryonic Germ (EG) Cells, and Embryonic Carcinoma (EC) Cells. These EC cells can be isolated from teratocarcinomas, a tumor that occasionally occurs in the gonad of a fetus. Unlike the other two, EC cells are usually aneuploid.
  • multipotent stem cells refers to true stem cells that can only differentiate into a limited number of types.
  • the bone marrow contains multipotent stem cells that give rise to all the cells of the blood but may not be able to differentiate into other cells types.
  • meenchymal stem cells refers to multipotent stromal cells that can differentiate into a variety of cell types, including osteoblasts, chondrocytes, and adipocytes (fat cells).
  • hematopoietic stem cell refers to a stem cell that is capable of differentiating into both myeloid lineages (i.e. monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets and some dendritic cells) and lymphoid lineages (i.e. T-cells, B-cells, NK-cells, and some dendritic cells).
  • myeloid lineages i.e. monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets and some dendritic cells
  • lymphoid lineages i.e. monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets and some dendritic cells
  • lymphoid lineages i.e. monocytes
  • the term "ingredient” refers to any compound, whether of chemical or biological origin, that can be used in cell culture media to maintain or promote the growth or proliferation of cells.
  • component e.g., fetal calf serum
  • ingredient can be used interchangeably and are all meant to refer to such compounds.
  • Typical ingredients that are used in cell culture media include amino acids, salts, metals, sugars, lipids, nucleic acids, hormones, vitamins, fatty acids, proteins and the like.
  • Other ingredients that promote or maintain growth of cells ex vivo can be selected by those of skill in the art, in accordance with the particular need.
  • cell culture is meant cells or tissues that are maintained, cultured or grown in an artificial, in vitro environment.
  • culture vessel refers to glass containers, plastic containers, or other containers of various sizes that can provide an aseptic environment for growing cells.
  • flasks, single or multiwell plates, single or multiwell dishes, or multiwell microplates can be used.
  • a bioreactor can be used to culture cells.
  • cell culture medium refers to a nutritive solution for culturing or growing cells.
  • contacting refers to the mixing, adding, seeding, or stirring of one or more cells with one or more compounds, solutions, media, etc.
  • a "serum-free" medium is a medium that contains no complete serum (e.g., human serum, fetal bovine serum, horse serum, goat serum, etc.).
  • buffering agent refers to an agent which acts to stabilize the hydrogen ion concentration and therefore the pH of a solution by neutralizing, within limits, both acids and bases.
  • Suitable buffering agents which can be used in the supplement and the medium of the present invention include N-[2-hydroxyethyl]piperazine-N'-[2- ethanesulfonic acid] (HEPES), ⁇ -glycerol-phosphate, and bicarbonate buffer.
  • subject refers to any animal, including mammals, birds, reptiles and amphibians and in preferred embodiments to mammals, including humans, companion animals, food production animals and wild animals.
  • compositions that are useful in methods of the present invention, such methods including in vivo methods for causing stem cell mobilization, homing and/or differentiation in a subject by administration of such compositions and ex vivo methods of expanding stem cells, providing stem cells to a subject, causing stem cells to differentiate,
  • Such compositions are useful in administration to a subject and indirect contact with stem cells or as an additive or supplement to cell culture medium.
  • the compositions of the present invention are particularly suited for supporting the expansion of mesenchymal stem cells (MSCs).
  • MSCs mesenchymal stem cells
  • Such cells include but are not limited to, chondrocytes, osteoclasts, osteoblasts, and epithelial cells of skin and vascular tissue.
  • compositions of the present invention is also suited for supporting the expansion of both primary and immortalized cells of most or all embryonic origin (e.g., ectodermal derivatives, mesodermal derivatives, and endodermal derivatives).
  • These cells include cells of the central and peripheral nervous system (neurons, glial cells, and astrocytes), epithelial cells (sensory epithelial cells, epidermal cells (skin, mammary, hair, nails, pituitary gland, sebaceous gland)), connective tissue cells (cartilage, bone, striated and smooth muscle, hematopoietic cells, lymphoid cells, kidney, gonadal cells, adrenal cells) and parenchymal cells of the liver, pancreas, thyroid, thymus, as well as epithelial linings of the urinary bladder, urethra, tympanic cavity, and eustachian tube.
  • These cells can be of human or other mammalian or eukaryotic origin
  • compositions of the invention include, aspartyl-alanyl diketopiperazine (i . e . , "Asp -Al a D KP or "DA-DKP").
  • Diketopiperazines (DKP) are a class of cyclic organic compounds that result from peptide bonds between two amino acids to form a lactam. They are the smallest possible cyclic peptides.
  • the invention also provides for a pharmaceutical product comprising a DA-DKP composition.
  • diketopiperazines such as DA-DKP
  • these methods may be employed to synthesize the diketopiperazines of the invention. See, e.g., U.S. Patents Nos. 4,694,081, 5,817,751, 5,990,112, 5,932,579 and 6,555,543, US Patent Application Publication Number 2004/0024180, PCT applications WO 96/00391 and WO 97/48685, and Smith et al, Bioorg. Med. Chem. Letters, 8, 2369-2374 (1998), the complete disclosures of which are incorporated herein by reference.
  • diketopiperazines such as DA-DKP
  • DA-DKP can be prepared by first synthesizing dipeptides.
  • the dipeptides can be synthesized by methods well known in the art using L-amino acids, D-amino acids or a combination of D- and L-amino acids.
  • solid-phase peptide synthetic methods can be used.
  • dipeptides are also available commercially from numerous sources, including DMI Synthesis Ltd., Edinburgh, UK (custom synthesis), Sigma-Aldrich, St. Louis, MO (primarily custom synthesis), Phoenix Pharmaceuticals, Inc., Belmont, CA (custom synthesis), Fisher Scientific (custom synthesis) and Advanced ChemTech, Louisville, KY.
  • the dipeptide is synthesized or purchased, it is cyclized to form a diketopiperazine. This can be accomplished by a variety of techniques.
  • U.S. Patent Application Publication Number 2004/0024180 describes a method of cyclizing dipeptides. Briefly, the dipeptide is heated in an organic solvent while removing water by distillation.
  • the organic solvent is a low-boiling azeotrope with water, such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n-butanol, 2-butanol, t-butanol, acetic acid butylester, carbon tetrachloride, chlorobenzene chloroform, cyclohexane, 1,2- dichlorethane, diethylacetal, dimethylacetal, acetic acid ethylester, heptane, methylisobutylketone, 3-pentanol, toluene and xylene.
  • water such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n-butanol, 2-butanol, t-butanol, acetic acid butylester, carbon tetrachloride, chlorobenzene chloroform, cyclohexane, 1,2- dich
  • the temperature depends on the reaction speed at which the cyclization takes place and on the type of azeotropic agent used.
  • the reaction is preferably carried out at 50-200°C, more preferably 80-150°C.
  • the pH range in which cyclization takes place can be easily determined by the person skilled in the art. It will advantageously be 2-9, preferably 3-7.
  • the dipeptide When one of the amino acids of the dipeptide has, or is derivatized to have, a carboxyl group on its side chain (e.g., aspartic acid), the dipeptide is preferably cyclized as described in U.S. Patent No. 6,555,543. Briefly, the dipeptide, with the side-chain carboxyl still protected, is heated under neutral conditions. Typically, the dipeptide will be heated at from about 80°C to about 180°C, preferably at about 120°C. The solvent will be a neutral solvent.
  • the solvent may comprise an alcohol (such as butanol, methanol, ethanol, and higher alcohols, but not phenol) and an azeotropic co-solvent (such as toluene, benzene, or xylene).
  • an alcohol such as butanol, methanol, ethanol, and higher alcohols, but not phenol
  • an azeotropic co-solvent such as toluene, benzene, or xylene.
  • the alcohol is butan-2-ol
  • the azeotropic co-solvent is toluene.
  • the heating is continued until the reaction is complete, and such times can be determined empirically.
  • the dipeptide will be cyclized by refluxing it for about 8-24 hours, preferably about 18 hours.
  • the protecting group is removed from the diketopiperazine.
  • strong acids mineral acids, such as sulfuric or hydrochloric acids
  • strong bases alkaline bases, such as potassium hydroxide or sodium hydroxide
  • strong reducing agents e.g., lithium aluminum hydride
  • Dipeptides made on solid phase resins can be cyclized and released from the resin in one step. See, e.g., U.S. Patent No. 5,817,751.
  • the resin having an N- alkylated dipeptide attached is suspended in toluene or toluene/ethanol in the presence of acetic acid (e.g., 1%) or triethylamine (e.g., 4%).
  • acetic acid e.g., 1
  • triethylamine e.g., 4%
  • basic cyclization conditions are preferred for their faster cyclization times.
  • diketopiperazines Other methods of cyclizing dipeptides and of making diketopiperazines are known in the art and can be used in the preparation of diketopiperazines useful in the practice of the invention. See, e.g., those references listed above.
  • many diketopiperazines suitable for use in the present invention can be made as described below from proteins and peptides.
  • diketopiperazines for use in the practice of the invention can be obtained commercially from, e.g., DMI Synthesis Ltd., Edinburgh, UK (custom synthesis).
  • the DA-DKP composition and/or products of the present invention can be prepared from solutions containing DA-DKP, including from the commercially-available pharmaceutical compositions comprising albumin, such as human serum albumin. It has been found that DA-DKP is present in some commercially-available intravenous pharmaceutical compositions containing albumin.
  • the DA-DKP present in these pharmaceutical preparations is formed by the heating steps often used in the manufacture of these pharmaceutical compositions. The heating results in cleavage and cyclization of the two N-terminal amino acids of the proteins to form DA-DKP.
  • DA-DKP can be prepared by heating solutions of albumin as well as other proteins and peptides.
  • a solution of albumin in phosphate buffer at neutral pH is prepared.
  • the solution is a concentrated solution (e.g., about 100- 500 mM) to achieve protonation of the N-terminal amino acid.
  • the solution is heated at 60°C for from about 2 hours to several days, preferably about 4 days, to cause formation of DA-DKP.
  • Denaturation of the protein should, preferably, be avoided. This can be accomplished by using shorter times and/or by adding caprylic acid or N-acetyl tryptophan at about 0.02 M for each.
  • DA-DKP can also be prepared by contacting a solution of albumin with an enzyme that can cleave the two N-terminal amino acids from the protein or peptide (e.g., dipeptidyl peptidases, particularly DPP IV). Suitable dipeptidyl peptidases are available commercially from, e.g., Sigma.
  • the reaction should be conducted at pH 6-8, preferably in a buffer, such as phosphate buffer, at a temperature high enough to speed the reaction but not so high that the protein is denatured (e.g., 37°C).
  • Preparation of the DA-DKP compositions can be by well known methods, such as ultrafiltration, chromatography (e.g., size-exclusion chromatography), affinity chromatography (e.g., using a column of beads having attached thereto an antibody or antibodies directed to the desired diketopiperazine(s) or an antibody or antibodies directed to the truncated protein or peptide), anion exchange or cation exchange, sucrose gradient centrifugation, chromatography, salt precipitation, or sonication, that will remove some or all of the albumin in the solution.
  • the resultant DA-DKP-containing composition and/or product can be used and incorporated into pharmaceutical compositions as described above.
  • a composition of the invention is prepared by treating an albumin-containing feed stream by tangential fiow filtration.
  • tangential fiow refers to the direction of fiow of the albumin-containing feed stream relative to the filtration media. This flow direction may be either tangential (also commonly referred to as “cross flow”), or "normal flow", or a combination of both. Tangential flow refers to an albumin-containing feed stream characterized by most of the stream flowing across the filtration media surface, whereas normal fiow refers to a stream characterized by most of the stream flowing through the filtration media, at a 90° angle relative to the filtration media surface.
  • a composition of the invention is produced by treating an albumin-containing feed stream by chromatography.
  • Reference herein to chromatography is to the mechanical and/or physical operation of separating one fraction of the albumin- containing feed stream from the remaining fraction by use of a pressure drop across a stationary phase.
  • mechanical chromatography refers to, but is not limited to, size exclusion chromatography.
  • physical chromatography refers to, but is not limited to, affinity chromatography, ion exchange chromatography, fast protein liquid chromatography and immunoaffinity chromatography.
  • the stationary phase of a chromatography step may include, but is not limited to, resins (i.e. polystyrene, polystyrene divinylbenzene and polyacrylamide), ion exchange resins (i.e. sulfonated, quaternary ammonium, carboxylate and diethyl ammounium functional groups), cross-linked agarose, cross-linked dextrans, phosphocellulose, porous glass and silica, alumina and zirconia matrices.
  • the stationary phase may be immobilized on a solid support particle, or on the inner wall of a cylinder, either by physical attraction, chemical bonding, and or by in situ polymerization after coating.
  • the immobilized stationary phase may coat the outer surfaces of the particles and cylinder, and/or fill any available pores within the solid particles.
  • the stationary phase may be functionalized with biospecific ligands which include, but is not limited to, antibodies, protein receptors, steroid hormones, vitamins and enzyme inhibitors.
  • a human serum albumin composition can be passed over an ultrafiltration membrane having a molecular weight cut-off that retains the albumin while the DA-DKP passes into the resulting filtrate or fraction.
  • This filtrate may comprise components having molecular weights less than about 50 kDA, less than about 40 kDa, less than 30 kDa, less than about 20 kDa, less than about 10 kDa, less than about 5 kDa, less than about 3 kDa.
  • the filtrate comprises components having molecular weights less than about 5 Da (also referred to as " ⁇ 5000MW").
  • This ⁇ 5000MW fraction or filtrate contains DA-DKP which is formed after the dipeptide aspartate-alanine is cleaved from albumin and subsequently cyclized into the diketopiperazine.
  • Physiologically-acceptable salts of the DA-DKP of the invention may also be used in the practice of the invention.
  • Physiologically-acceptable salts include conventional non-toxic salts, such as salts derived from inorganic acids (such as hydrochloric, hydrobromic, sulfuric, phosphoric, nitric, and the like), organic acids (such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, glutamic, aspartic, benzoic, salicylic, oxalic, ascorbic acid, and the like) or bases (such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation or organic cations derived from ⁇ , ⁇ -dibenzylethylenediamine, D-glucosamine, or ethylenediamine).
  • the salts are prepared in a conventional manner, e.g., by neutralizing the free base form of the compound with an
  • the composition of the present invention can be a pharmaceutical solution having a DA-DKP concentration range with a lower endpoint of about 10 ⁇ , about 20 ⁇ , about 30 ⁇ , about 40 ⁇ , about 50 ⁇ , about 60 ⁇ , about 70 ⁇ , about 80 ⁇ , about 90 ⁇ , about 100 ⁇ , about 110 ⁇ , about 120 ⁇ , about 130 ⁇ , about 140 ⁇ , about 150 ⁇ , about 160 ⁇ , about 170 ⁇ , about 180 ⁇ , about 190 ⁇ , about 200 ⁇ , about 210 ⁇ , about 220 ⁇ , about 230 ⁇ , about 240 ⁇ , about 240, about 250 ⁇ , about 260 ⁇ , about 270 ⁇ , about 280 ⁇ , about 290 ⁇ , about 300 ⁇ , about 310, about 320 ⁇ , about 330 ⁇ , about 340 ⁇ , about 350 ⁇ , about 360 ⁇ , about 370 ⁇ , about 380 ⁇ , about 390 ⁇ , or about 400 ⁇ .
  • the composition of the present invention may be a pharmaceutical solution having a DA-DKP concentration range with an upper endpoint of about 600 ⁇ , about 580 ⁇ , about 570 ⁇ , about 560 ⁇ , about 550 ⁇ , about 540 ⁇ , about 530 ⁇ , about 520 ⁇ , about 510 ⁇ , about 500 ⁇ , about 490 ⁇ , about 480 ⁇ , about 470 ⁇ , about 460 ⁇ , about 450 ⁇ , about 440 ⁇ , about 430 ⁇ , about 420 ⁇ , about 410 ⁇ , about 400 ⁇ , about 390 ⁇ , about 380 ⁇ , about 370 ⁇ , about 360 ⁇ , about 350, about 340 ⁇ , about 330 ⁇ , about 320 ⁇ , about 310 ⁇ , about 300 ⁇ , about 290 ⁇ , about 280, about 270 ⁇ , about 260 ⁇ , about 250 ⁇ , about 240 ⁇ , about 230 ⁇ , about 220 ⁇ , about 210 ⁇ , or about
  • An effective amount of DA-DKP in the composition of the present invention for treating conditions described herein can be a range with a lower endpoint of about 10 ⁇ g, about 15 ⁇ g, about 20 ⁇ g, about 25 ⁇ g, about 30 ⁇ g, about 35 ⁇ g, about 40 ⁇ g, about 45 ⁇ g, about 50 ⁇ g, about 55 ⁇ g, about 60 ⁇ g, about 65 ⁇ g, about 70 ⁇ g, about 75 ⁇ g, about 80 ⁇ g, about 85 ⁇ g, about 90 ⁇ g, about 95 ⁇ g, about 100 ⁇ g, about 110 ⁇ g, about 120 ⁇ g, about 130 ⁇ g, about 140 ⁇ g, about 150 ⁇ g, about 160 ⁇ g, about 170 ⁇ g, about 180 ⁇ g, about 190 ⁇ g, about 200 ⁇ g, about 210 ⁇ g, about 220 ⁇ g, about 230 ⁇ g, about 240 ⁇ g, about 250 ⁇ g, about 260 ⁇ g, about
  • an effective amount of DA-DKP in the composition of the present invention for treating conditions described herein can be a range with upper endpoint of about 500 ⁇ g, about 490 ⁇ g, about 480 ⁇ g, about 470 ⁇ g, about 460 ⁇ g, about 450 ⁇ g, about 440 ⁇ g, about 430 ⁇ g, about 420 ⁇ g, about 410 ⁇ g, about 400 ⁇ g, about 390 ⁇ g, about 380 ⁇ g, about 370 ⁇ g, about 360 ⁇ g, about 350 ⁇ g, about 340 ⁇ g, about 330 ⁇ g, about 320 ⁇ g, about 310 ⁇ g, about 300 ⁇ g, about 290 ⁇ g, about 280 ⁇ g, about 270 ⁇ g, about 260 ⁇ g, about 250 ⁇ g, about 240 ⁇ g, about 230 ⁇ g, about 220 ⁇ g, about 210 ⁇ g, about 200 ⁇ g, about 190 ⁇ g, about 180
  • Dosage forms for the topical or transdermal administration of compounds of the invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and drops.
  • the active ingredient may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to the active ingredient, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to the active ingredient, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder or mixtures of these substances.
  • Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of compounds of the invention to the body.
  • dosage forms can be made by dissolving, dispersing or otherwise incorporating one or more compounds of the invention in a proper medium, such as an elastomeric matrix material.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate-controlling membrane or dispersing the compound in a polymer matrix or gel.
  • compositions of this invention suitable for parenteral administrations comprise one or more compounds of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • These compositions may also contain adjuvants such as wetting agents, emulsifying agents and dispersing agents.
  • isotonic agents such as sugars, sodium chloride, and the like in the compositions.
  • prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monosterate and gelatin.
  • the absorption of the drug in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. The injectable materials can be sterilized for example, by filtration through a bacterial-retaining filter.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use.
  • sterile liquid carrier for example water for injection
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the type described above.
  • kits comprising the pharmaceutical products of the present invention are also provided.
  • the kits can comprise a DA-DKP composition formulated for administration by injection.
  • the DA-DKP can be prepared as described herein, such as by removing albumin from a solution of a human albumin composition.
  • the kits may contain unit- dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use.
  • the kits may also be stored in a condition, wherein the contents are ready for direct use or injection. While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition).
  • compositions of the invention comprise a compound or compounds of the invention as an active ingredient in admixture with one or more pharmaceutically-acceptable carriers and, optionally, with one or more other compounds, drugs or other materials.
  • Each carrier must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the animal.
  • Pharmaceutically-acceptable carriers are well known in the art. Regardless of the route of administration selected, the compounds of the present invention are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art. See, e.g., Remington's Pharmaceutical Sciences.
  • composition of the present invention may further comprise N-acetyl- tryptophan (NAT), caprylic acid, caprylate or combinations thereof.
  • the composition may comprise NAT.
  • Compositions of the present invention having NAT, caprylic acid, caprylate or combinations thereof may be a pharmaceutical composition having a NAT, caprylic acid, caprylate or combinations thereof in a concentration range with a lower endpoint of about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 11 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 16 mM, about 17 mM, about 18 mM, about 19 mM, or about 20 mM.
  • compositions of the present invention having NAT, caprylic acid, caprylate or combinations thereof may be a pharmaceutical composition having a NAT, caprylic acid, caprylate or combinations thereof in a concentration range with an upper endpoint of about 40 mM, about 39 mM, about 38 mM, about 37 mM, about 36 mM, about 35 mM, about 34 mM, about 33 mM, about 32 mM, about 31 mM, about 30 mM, about 29 mM, about 28 mM, about 27 mM, about 26 mM, about 25 mM, about 24 mM, about 23 mM, about 22, or about 21 mM.
  • the concentration range is about 4 mM to about 20 mM.
  • composition of the present invention may also comprise a second drug such as an analgesic (such as lidocaine or paracetoamol), an anti-inflammatory (such as bethamethasone, non-steroid anti-inflammatory drugs (NSAIDs), acetaminophen, ibuprofen, naproxen), and/or other suitable drugs.
  • analgesic such as lidocaine or paracetoamol
  • an anti-inflammatory such as bethamethasone, non-steroid anti-inflammatory drugs (NSAIDs), acetaminophen, ibuprofen, naproxen
  • NSAIDs non-steroid anti-inflammatory drugs
  • compositions comprising albumin found in the mammalian recipient of the treatments of this disclosure can be administered to stimulate stem cell expansion in the mammal.
  • compositions comprising these proteins and/or peptides which are currently available commercially can be used if they contain diketopiperazines, especially DA-DKP, it is preferred to treat the albumin as described above to increase the content of the DA-DKP before administration of the improved compositions.
  • the mammal is preferably a human, and the proteins and/or peptides are preferably human proteins and/or peptides. Parenteral routes of administration are preferred.
  • compositions or cell culture media supplemented with the compositions of the present invention the growth, expansion, and differentiation of stem cells can be regulated by the addition of defined growth factors or other cytokines. Such influence over stem cells in typical serum-containing culture is not possible as undefined components in serum obscure the cellular responses to defined factors.
  • stem cells can be expanded and differentiated in suspension culture and in the absence of stromal cells, collagen, or support matrices.
  • the supplement and compositions or cell culture media supplemented with the compositions of the present invention provide for the growth and expansion of both pluripotent stem cell populations and differentiated progeny.
  • compositions or cell culture media supplemented with the compositions of the present invention can be used to culture stem cells derived from a number of animals, including human, monkey, ape, mouse, rat, hamster, rabbit, guinea pig, cow, swine, dog, horse, cat, goat, and sheep.
  • human stem cells are cultured.
  • the stem cells are totipotent, pluripotent or multipotent stem cells.
  • the stem cells are embryonic stem cells, fetal stem cells, extraembryonic stem cells or adult stem cells.
  • compositions or cell culture media supplemented with the compositions of the present invention may also include one or more ingredients selected from the group consisting of one or more antioxidants, one or more albumins or albumin substitutes, one or more lipid agents, one or more insulins or insulin substitutes, one or more transferrins or transferrin substitutes, one or more trace elements, one or more glucocorticoids, N- acetyl-L cysteine, Human Ex-Cite®, ethanolamine, human zinc insulin, human iron saturated transferrin, selenium, hydrocortisone, D,L-tocopherol acetate, and 2- mercaptoethanol.
  • N-acetyl-L-cysteine can be solubilized in double-distilled water.
  • the pH of the N-acetyl-L-cysteine at this point is approximately 2.0.
  • the pH of the N-acetyl-L-cysteine is adjusted to 7.0 with 5N sodium hydroxide and is added to the mixture. The entire mixture is then filtered through a 0.2 micron low protein binding filter.
  • the supplement or the medium of the present invention can be in liquid form or can be maintained in dry form.
  • the type of liquid carrier and the method used to dissolve the ingredients into solution vary and can be determined by one of ordinary skill in the art with no more than routine experimentation.
  • the liquid carrier is water.
  • the supplement or the medium or concentrated formulation of the present invention are typically sterilized to prevent unwanted contamination. Sterilization may be accomplished, for example, by ultraviolet light, filtration, or heat.
  • ingredients of the supplement of the present invention which are of human origin (e.g., human serum albumin, transferrin) are heat treated, prior to use, by heating at 60° C. for 10 hours.
  • human origin e.g., human serum albumin, transferrin
  • the present invention also provides a kit comprising a carrier means such as a box or carton being compartmentalized to receive in close confinement therein one or more container means such as vials, tubes, ampules, jars, and the like, wherein a first container means contains the compositions or cell culture media supplemented with the compositions of the present invention.
  • a second container means contains a basal medium.
  • the container containing the supplement of the present invention can be stored from about -135 to about 4°C, preferably from about -5 to about -80°C, most preferably from about -5 to about -20°C, and still more preferably at about -20°C.
  • a container containing the medium of the invention is preferably stored at about 2 to about 8°C, and most preferably at about 4°C.
  • the present invention also provides a composition comprising stem cells in a serum-free medium, wherein the serum-free medium, which is supplemented with compositions of the present invention, which is capable of supporting the growth of stem cells in serum-free culture. Aliquots of this composition can be frozen at about -80°C. and below. Aliquots of this composition can be stored indefinitely at less than or equal to about -135°C. After an aliquot of the composition has been thawed and opened, using sterile cell culture technique, the stem cells can be cultivated in serum-free culture.
  • compositions of the invention in storage and transportation of stem cells in order to maintain viability, mobility and stem cell function.
  • a composition of the invention may be used alone or added to a variety of agents known in the art to allow transportation of stem cells. This is particularly important in situations of bone marrow stem cell transportation in which cell freezing and thawing is not performed in numerous situations. The ability to adequately store stem cells during transportation would allow for tissue extraction at separate physical locations from the stem cell processing facility.
  • compositions of the invention are a pharmaceutical preparation comprising a composition of the invention generated in a Good Manufacturing Practices/Good Tissue Practices environment such that it is suitable for clinical use. Subsequent to concentration and quantification of units of activity, the composition of the invention can be diluted into an excipient or carrier. It may be advantageous to formulate compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for an individual to be treated; each unit containing a predetermined quantity of active compounds of the compositions of the invention calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the dosage unit forms of the invention are dependent upon the amount of a composition necessary to stimulate proliferation of the respective stem cells whose proliferation and/or differentiation is being sought.
  • the amount of composition necessary to stimulate proliferation and/or differentiation of the desired stem cells can be formulated in a single dose, or can be formulated in multiple dosage units. Treatment can require a one-time dose, or can require repeated doses.
  • compositions of the invention will be performed in agreement with standard practices that are known to one skilled in the art. These are well known in the art and the one chosen is based upon the route of administration that will be used, as well as specific pharmacokinetic properties that are desired.
  • the preferred embodiment of therapy utilizing a composition of the invention is an injectable dosage, and more preferably, an injection formulated for administration into the specific area requiring regeneration of stem cells.
  • injectable dosage and more preferably, an injection formulated for administration into the specific area requiring regeneration of stem cells.
  • several embodiments are possible.
  • routes of administration can include parenteral, e.g, intra-articular injection, intravenously, intradermally, intraspinally, intraperitoneally, subcutaneously, or intramuscularly, oral (e.g., ingestion or inhalation), transdermal (topical), transmucosal, and rectal administration.
  • parenteral e.g, intra-articular injection, intravenously, intradermally, intraspinally, intraperitoneally, subcutaneously, or intramuscularly, oral (e.g., ingestion or inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions for formulating therapeutic compositions of the invention can include: sterile diluent such as water for injection, saline solution (e.g., phosphate buffered saline (PBS, UPS)), fixed oils, glycerine, or other synthetic solvents; antibacterial and antifungal agents such as parabens, a polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), chlorobutanol, phenol, ascorbic acid, thimerosal, and the like; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • sterile diluent such as water for injection, saline solution (e.g., phosphate buffered saline (
  • the desired fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • a coating such as lecithin
  • surfactants for example, sugars, sodium chloride, polyalcohols such as mannitol or sorbitol, and in the composition.
  • Prolonged administration of the injectable compositions can be brought about by including an agent that delays absorption.
  • agents include, for example, aluminum monostearate and gelatin.
  • the parenteral preparation can be enclosed in ampules, disposable syringes, or multiple dose vials made of glass or plastic.
  • Oral compositions can be liquid, or can be enclosed in gelatin capsules or compressed into tablets. Tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose; a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; colloidal silicon dioxide.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • Embodiments of the invention include the use of the compositions of the invention to effect the mobilization, homing, differentiation and/or expansion of stem cells within a living organism.
  • Clinical situations where administration of the compositions of the invention is desirable include conditions where an increase in the number of stem cells is sought due to disease or senescence of endogenous stem cells or conditions benefiting from reparative function on the surrounding tissues or elsewhere in the organism.
  • Such stem cells can be present in pancreatic tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone marrow tissue, bone spongy tissue, cartilage tissue, liver tissue, pancreas tissue, pancreatic ductal tissue, spleen tissue, thymus tissue, Peyer's patch tissue, lymph nodes tissue, thyroid tissue, epidermis tissue, dermis tissue, subcutaneous tissue, heart tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophagus tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterus tissue, eye tissue (including retinal tissue), lung tissue, testicular tissue, ovarian tissue, prostate tissue, connective tissue, endocrine tissue, and mesentery tissue.
  • compositions of the invention can be performed systemically, or in a localized environment in the subject.
  • a site of local administration can be any site in the body in which the development of tissue is desired or beneficial, such as a joint, a surgical site, a site of a segmented skeletal gap or non-union fracture, a wound, an ulcer, or an inflammatory skin rash.
  • the compositions of the invention can also be administered systemically, such as by an oral dosage formulation.
  • compositions of the invention can be administered to a subject in, on or as part of an implantable device.
  • such a device can be a sponge, biocompatible polymer, bioerodible polymer, putty, gel, bone matrix, artificial bone matrix, bolt, screw, endotracheal tube, stent, contact lens, pacemaker, central IV tube, foley catheter, or intracranial device.
  • the invention includes a method to cause an effect selected from stem cell mobilization, stem cell homing, stem cell expansion, and stem cell differentiation in a subject by administering DA-DKP to a subject that has a need therefor.
  • the step of administering DA-DKP can have an effect of increasing the production of one or more of CXCR4, CXCL12, MMP13, MMP14, aggrecan, SDF1, and collagen 2A1.
  • the step of administering DA-DKP can have an effect of decreasing production of one or more proteins selected from MAPK-activated protein kinase 3, beta-adrenergic receptor kinase 1, ADAM metallopeptidase with thrombodpondin type I motif, MAPK- activated protein kinase 2, C-Src kinase, Macrophage Scavenger Receptor, Noggin, Tyrosine kinase Bruton, Glycogen synthase kinase-3 alpha/beta, Glycogen synthase kinase-3 alpha/beta, HSP 90 alpha/beta, HSP 90 alpha/beta, Phosphoinositide-3 -kinase, catalytic subunit alpha, and Eukaryotic translation initiation factor 4A, and Fibroblast Growth Factor 17, and in particular, one or more proteins selected from MAPK-activated protein kinase 3, Nogg
  • the step of administering DA-DKP can have an effect of increasing production of one or more proteins selected from Clusterin (Apolipoprotein J), Prothrombin, C1QBP (Hyaluronan binding protein 1), TNFSF 15 (VEGF inhibitor), Mammaglobin 2, MIP3b (CCL 19), MCP 1 (CCL 2), PTHrP, Spondin 1, Elafm (elastase inhibitor), IL 11, NPS- PLA2, CFC 1 (cryptic protein), Testican 1 (SPOCK 1), Angiogenin, URB, MMP-3, IP10 (cxcl 10), BSSP 4, IL 8 (cxcl 8), RSP02, Cystatin C, bFGF, Factor H, Coagulation Factor IX, SDF-1 (cxcl 12), CATC (Dipeptidyl peptidase 1), PIGR, Ck-b-8-1 (MPIF 1 splice variant), Cls, EMR2, ART, DPP 2,
  • the administration of DA-DKP can decrease the production of a protein selected from MAPK-activated protein kinase 3, Noggin, Phosphoinositide-3-kinase, catalytic subunit alpha, and combinations thereof and increase the production of a protein selected from Clusterin (Apolipoprotein J), C1QBP (Hyaluronan binding protein 1), MCP 1 (CCL 2), PTHrP, Elafm (elastase inhibitor), IL 11, MMP-3, bFGF, SAA, TIMP-1, Semaphorin 3 A, and combinations thereof.
  • a protein selected from MAPK-activated protein kinase 3, Noggin, Phosphoinositide-3-kinase, catalytic subunit alpha, and combinations thereof and increase the production of a protein selected from Clusterin (Apolipoprotein J), C1QBP (Hyaluronan binding protein 1), MCP 1 (CCL 2), PTHrP, Elafm (elast
  • Methods of the invention include methods for stimulating development of tissue in a subject by administering DA-DKP to a subject. These methods are suitable for the development of any tissue in the subject, including one or more of nervous system tissue, adipose tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone spongy tissue, cartilage tissue, pancreatic ductal tissue, spleen tissue, thymus tissue, tonsil tissue, Peyer's patch tissue, lymph nodes tissue, thyroid tissue, epidermis tissue, dermis tissue, subcutaneous tissue, heart tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophagus tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterus tissue, eye tissue, lung tissue, testicular tissue, ovarian tissue, prostate tissue, connect
  • a specific in vivo method of treatment includes conditions in which a higher number and/or more rapid recovery of stem cells is needed in a subject after a medical procedure.
  • a bone marrow transplant expansion of hematopoietic cells is desirable in order for the patient not to succumb to bacterial or viral infections.
  • Such expansion of granulocytic and monocytic precursors would be useful in enhancing immunological defenses subsequent to a bone marrow transplant. Accordingly, this invention provides methods and compositions that can be administered to a patient having undergone a bone marrow transplant.
  • compositions of the invention to patients having an injury in a tissue selected from pancreatic tissue, liver tissue, smooth muscle tissue, striated muscle tissue, cardiac muscle tissue, bone tissue, bone marrow tissue, bone spongy tissue, cartilage tissue, liver tissue, pancreas tissue, pancreatic ductal tissue, spleen tissue, thymus tissue, Peyer's patch tissue, lymph nodes tissue, thyroid tissue, epidermis tissue, dermis tissue, subcutaneous tissue, heart tissue, lung tissue, vascular tissue, endothelial tissue, blood cells, bladder tissue, kidney tissue, digestive tract tissue, esophagus tissue, stomach tissue, small intestine tissue, large intestine tissue, adipose tissue, uterus tissue, eye tissue (including
  • Another embodiment of the invention is the administration of a composition of the invention alone or in combination with growth factors to promote healing.
  • Another embodiment of the invention is a method of treatment for diabetes by administering compositions of the invention to a subject with diabetes to induce islet regeneration.
  • This method can further include administration of other factors capable of inducing islet regeneration.
  • factors can be, for example, soluble proteins, membrane bound proteins, or intracellular acting transcription factors.
  • GLP-1, EGF and gastrin administration into mice combinations of GLP-1, EGF and gastrin leads to regeneration of islets or islet-like cells that are functionally effective in models like NOD or streptozocin induced diabetes.
  • compositions of the invention can be added to cultures of differentiating islets in vitro to expand the stem cell numbers, but can also be administered directly to the pancreas of a patient in vivo in order to restore islet cell function or to amplify the effect of administered hormones.
  • an in vivo method of the invention is a treatment for multiple sclerosis by administering a composition of the invention to a patient having multiple sclerosis for expanding neuronal progenitor cells.
  • a composition of the invention can be administered to patients suffering from multiple sclerosis alone or in combination with agents capable of inhibiting the autoimmune process. Synergy in therapeutic effects is anticipated through the concurrent induction of tissue healing and immune system repair.
  • Another embodiment of the invention is the treatment of an immunological disorder, such as an autoimmune disorder, by extracting stem cells from an autologous patient, treating the cells with a composition of the invention and/or other combinations of stem cell-expanding compounds, ablating the immune system of the patient, and subsequent reintroduction of stem cells into the host for reconstitution.
  • a composition of the invention may be subsequently provided directly to the host in order to accelerate reconstitution of hematopoiesis.
  • Autoimmune diseases treatable by these procedures include, but are not limited to, Type 1 diabetes, multiple sclerosis, rheumatoid arthritis, systemic sclerosis, Hashimoto's thyroiditis, myasthenia gravis, scleroderma, systemic lupus erythromatosis, graft versus host disease, and the like.
  • Another embodiment of the invention is a treatment for an autoimmune disorder such as Type 1 diabetes, multiple sclerosis, rheumatoid arthritis, Hashimoto's thyroiditis, myasthenia gravis, scleroderma, and the like comprising administering a composition of the invention to a relevant location within the organism in need of such treatment to enhance the production of stem cells, such as administration into a joint or into the pancreas of an organism to enhance chondrogenesis or islet cell production.
  • an autoimmune disorder such as Type 1 diabetes, multiple sclerosis, rheumatoid arthritis, Hashimoto's thyroiditis, myasthenia gravis, scleroderma, and the like
  • administering a composition of the invention to a relevant location within the organism in need of such treatment to enhance the production of stem cells, such as administration into a joint or into the pancreas of an organism to enhance chondrogenesis or islet cell production.
  • Another embodiment of the invention includes the use of a composition of the invention for expansion of antigen-specific and/or non-specific immune regulatory cells within an organism. Expansion of such cells is use for controlling pathological immune responses. For example, it is known that Th2 cells, TR1 cells, CD4+CD25+FoxP3+ cells, and CD3+ double negative cells, are capable of suppressing immune responses in an antigen specific manner, whereas NKT cells, myeloid suppressor cells, M2 cells, and immature dendritic cells are capable of suppressing immune responses in an antigen nonspecific manner.
  • a composition of the invention may be used for ex vivo culture and expansion of immune regulatory cells derived from a patient in need thereof.
  • a composition of the invention may be used either alone or in combination with factors known to be involved in the development of said cells for administration to an organism to enhance proliferation of these cells without loss of function.
  • Another embodiment of the invention is a treatment for stroke by administration of a composition of the present invention to a stroke patient to cause the in vivo mobilization, homing, expansion and/or differentiation of endogenous neural cells.
  • This method can further include administration of a composition of the invention in combination with polypeptides and proteins known in the art to expand neural cells.
  • Another embodiment of the invention is the use of a composition of the invention as an adjuvant to treatment for a degenerative condition by the application of a combination of a composition of the invention with known therapies in order to enhance the beneficial effects of known therapies.
  • Another embodiment of the invention relates to the use of a composition of the invention described herein in the preparation of a medicament for enhancing the expansion of stem cells in vivo or ex vivo.
  • a further embodiment of the invention is an in vivo method for the treatment of a degenerative joint disease by administration of a composition of the invention to cause stem cell mobilization, homing, expansion and/or differentiation in the subject.
  • a degenerative joint disease is a gradual deterioration of the articular cartilage that covers joints.
  • a degenerative joint disease (osteoarthritis) is a noninfectious progressive disorder of the weightbearing joints.
  • the normal articular joint cartilage is smooth, white, and translucent. It is composed of cartilage cells (chondrocytes) imbedded in a spongelike matrix made of collagen, protein polysaccharides, and water. With early primary arthritis, the cartilage becomes yellow and opaque with localized areas of softening and roughening of the surfaces.
  • cartilage As degeneration progresses, the soft areas become cracked and worn, exposing bone under the cartilage. The bone then begins to remodel and increase in density while any remaining cartilage begins to fray. Eventually, osteophytes (spurs of new bone) covered by cartilage form at the edge of the joint. As mechanical wear increases, the cartilage needs repairing. The cartilage cells are unable to produce enough of the sponge-like matrix and therefore the damaged cartilage cannot repair itself. The cartilage has no blood supply to enhance healing. The majority of degenerative joint disease is the result of mechanical instabilities or aging changes within the joint. This includes old age degenerative arthritis and, in younger individuals, may be the result of injuries, bruises, abnormal joint configuration (i.e.
  • hip dysplasia or mechanical wear from anterior cruciate ligament rupture, patellar luxation, or osteochondritis dissecans, for example.
  • Degenerative joint disease can occur at any joint in the body, including without limitation, knee, hip, shoulder, hand and spine.
  • Conventional pharmaceutical therapies for degenerative joint disease include administration of acetaminophen, nonsteroidal anti-inflammatory drugs (NSAIDS), narcotics, and corticosteroids.
  • NSAIDS nonsteroidal anti-inflammatory drugs
  • narcotics narcotics
  • corticosteroids corticosteroids
  • a particular embodiment of the invention is a method of stimulating chondrogenesis in a subject by administering a pharmaceutical composition that includes DA-DKP.
  • the pharmaceutical composition can include any pharmaceutical composition described herein and therefore, can also include a component selected from N-acetyl tryptophan, caprylate, caprylic acid, and combinations.
  • chondrogenesis can be stimulated in a stem cell, and the stem cell can be a progenitor cell or a mesenchymal stem call (MSC).
  • the stimulation of chondrogenesis in this method can promote cartilage, bone, and/or ligament repair or induce repair or regeneration of chondral tissue in the subject.
  • the method is useful for treating or ameliorating a chondrogenic disease, such as a congenital cartilage disease, degenerative or fibrotic joint, rheumatoid arthritis or osteoarthritis, in the subject.
  • a chondrogenic disease such as a congenital cartilage disease, degenerative or fibrotic joint, rheumatoid arthritis or osteoarthritis
  • the chondrogenesis can be useful to treat or repair a condition selected from a cartilage defect, a skeletal defect, and a fracture arising from trauma or surgery.
  • a skeletal defect can be a large segmental skeletal gap
  • the cartilage defect can be an articular cartilage tear, a congenital cartilage defect or cartilage damage induced by bone fractures.
  • This method can be conducted by stimulating stem cells ex vivo and then administering the stimulated stem cells to the subject.
  • the stem cells can be stimulated ex vivo by culturing a population of stem cells of chondrocyte lineage with DA- DKP, or composition comprising a DA-DKP, for a time sufficient to stimulate chondrogenesis, and then, be implanted into a desired site in the subject.
  • a further embodiment of the present invention includes a method of stimulating development of nervous tissue in a subject by administering a pharmaceutical composition that includes DA-DKP.
  • the pharmaceutical composition can include any pharmaceutical composition described herein and therefore, can also include a component selected from N-acetyl tryptophan, caprylate, caprylic acid, and combinations thereof to a subject.
  • the development of nervous tissue can be stimulated in a stem cell, which can be selected from a progenitor cell and mesenchymal stem call (MSC).
  • MSC mesenchymal stem call
  • the stimulation of development of nervous tissue can promote brain, spinal cord, and/or peripheral nerve repair or induces repair or regeneration of neurons, neuroglia, and/or astrocytes in the subject.
  • the development of nervous tissue can treat or ameliorate a disease of the central nervous system and/or a disease of the peripheral nervous system in the subject. Such diseases can be selected from neurodegenerative diseases.
  • the development of nervous tissue can also treat or repair a condition selected from an injury arising from trauma or surgery.
  • This method can be conducted by stimulating stem cells ex vivo and then administering the stimulated stem cells to the subject.
  • the stem cells can be stimulated ex vivo by culturing a population of stem cells of neuron, neuroglia, and/or astrocyte lineage with the pharmaceutical composition, for a time sufficient to stimulate development of nervous tissue and then, be implanted into a desired site in the subject.
  • Ex vivo expansion of stem cells for systemic administration represents an effective treatment modality for disorders or diseases susceptible to stem cell treatment.
  • mesenchymal stem cells reduced both acute and chronic graft versus host disease in a patient suffering severe, grade IV graft versus host disease in the liver and gut subsequent to bone marrow transplant.
  • Administration of 2 million cells/kg on day 73 after bone marrow transplant lead to a long term remission of graft versus host disease (Le Blanc, et al., 2004, Lancet 363: 1439-1441), demonstrating that
  • the present invention further provides methods of increasing the expansion of stem cells ex vivo.
  • the method involves contacting the stem cells with a composition of the invention or mixing or incubating the stem cells with a growth medium that includes a composition of the invention to expand the stem cell population for subsequent administration, either locally at a needed site or systemically.
  • Such methods make possible the use of stem cell populations, such as mesenchymal stem cells, that expand relatively slowly and are therefore often not practical for widespread clinical use.
  • the compositions of the invention in addition to increasing the rate of stem cell proliferation, may also maintain the stem cells in an undifferentiated state. Stem cells reside in unique physiological niches, and while growing cells within mimics of such niches has been performed, the mimics of the stem cell niche are often unusable in clinical situations.
  • the current invention provides compositions for enhancing the expansion of stem cells that may recreate conditions similar to stem cell niches using approaches that are translatable into the clinical situation.
  • bone marrow is commonly used as a source of therapeutic stem cells for myocardial disease, angina, and hematopoietic cell transplant.
  • bone marrow in general contains a wide number of different stem cells in addition to the standard, well known, hematopoietic, CD34+ stem cell.
  • CD34- hematopoietic stem cells, mesenchymal stem cells, and myogenic precursor cells have all been found in bone marrow, in addition to T cells, B cells, and relatively high levels of CD4+ CD25+ T regulatory cells.
  • the stem cells can be grown in a culture medium containing a composition of the invention to increase the population of a heterogeneous mixture of cells, or a purified stem cell population, in order to increase the rate of expansion or growth of the stem cells when grown in culture.
  • stem cell expansion techniques began with work aimed at increasing the number of colonies formed on semisolid media. Early experiments used a variety of uncharacterized sera and conditioned media. For example, trophoblast cell line conditioned media, xenogeneic stromal cell conditioned media, supernatants from tumor cells, and healthy lymphocytes were used. Work was also performed towards designing serum free systems, using ingredients such as human transferrin and bovine insulin. The specific advantage of such systems is that hematopoietic stem cells could be expanded without concurrent differentiation.
  • the initial long term culture systems required the use of murine feeder (stromal) cells since human lines had certain disadvantages in terms of hematopoietic promoting activity. Numerous drawbacks existed to the use of murine feeder cell lines to maintain stem cell viability and proliferative potential. Due to this, an effort was made to overcome difficulties in growth of human derived feeder cells, and a variety of such cells have been developed.
  • Stem cells to be expanded by methods of the present invention can be isolated from any organ of any mammalian organism, by any means known to one of skill in the art.
  • the stem cells can be derived from embryonic or adult tissue.
  • One of skill of the art can determine how to isolate the stem cells from the particular organ or tissue of interest, using methods known in the art.
  • the stem cell populations can also be enriched using antibodies to other stem cell surface markers.
  • markers include, but are not limited to, FLK-1, AC133, CD34, c-kit, CXCR-4, Oct-4, Rex-1, CD9, CD13, CD29, CD44, CD166, CD90, CD105, SH-3, SH-4, TRA-1-60, TRA-1-81, SSEA-4, Sox-2, and the like.
  • One of skill in the art will be able to determine the specific cell marker useful for isolating stem cells from the desired tissue.
  • IMDM Iscove's modified Dulbecco's Media
  • DMEM DMEM
  • KO-DMEM DMEM/F12
  • RPMI 1640 McCoy's 5 A medium
  • minimum essential medium alpha medium a-MEM
  • F-12K nutrient mixture medium Kaighn's modification, F-12K
  • X-vivo 20
  • Stemline CCIOO, H2000, Stemspan, MCDB 131 Medium
  • Opti- MEM I Reduced Serum Media, Waymouth's MB 752/1 Media, Williams Media E, Medium NCTC- 109, neuroplasma medium, BGJb Medium, Brinster's BMOC-3 Medium, CMRL Medium, C0 2 -Independent Medium, Leibovitz's L
  • growth factors can be added as desired.
  • growth factors and other components that can be added include but are not limited to thrombopoietin (TPO), stem cell factor (SCF), IL-1, IL-3, IL-7, flt-3 ligand (flt- 3L), G-CSF, GM-CSF, Epo, FGF-1, FGF-2, FGF-4, FGF-20, IGF, EGF, NGF, LIF, PDGF, bone morphogenic proteins (BMP), activin-A, VEGF, forskolin, glucocorticords, and the like.
  • TPO thrombopoietin
  • SCF stem cell factor
  • IL-1 IL-3
  • IL-7 flt-3 ligand
  • flt- 3L flt-3 ligand
  • G-CSF G-CSF
  • GM-CSF Epo
  • FGF-1, FGF-2, FGF-4, FGF-20 Epo, FGF
  • the initial isolation media can contain either serum such as fetal calf, horse, or human serum, or more preferably, serum substitution components.
  • serum substitutes have included bovine serum albumin (BSA), insulin, 2- mercaptoethanol and transferrin (TF).
  • the stem cells can then be stored for a desired period of time, if needed.
  • Stem cell storage methods are known to those of skill in the art.
  • the stem cells are treated to a cryoprotection process, then stored frozen until needed.
  • the stem cells can be purified prior to contact with a composition of the invention by methods known in the art, using, for example, antibody technology such as panning of cells, through the use of fluorescence activated cell sorting (FACS) methods, or magnet activated cell sorting methods such as that MACS apparatus, to isolate cells having the desired stem cell markers, or to remove unwanted, contaminating cell types having unwanted cell markers prior to contacting with compositions of the invention.
  • FACS fluorescence activated cell sorting
  • MACS magnet activated cell sorting
  • Other methods of stem cell purification or concentration can include the use of techniques such as counterflow centrifugal elutriation, equilibrium density centrifugation, velocity sedimentation at unit gravity, immune rosetting and immune adherence, T lymphocyte depletion.
  • stem cell markers examples include, but are not limited to, FLK-1, AC133, CD34, c-kit, CXCR-4, Oct-4, Rex-1, CD9, CD13, CD29, CD44, CD166, CD90, CD105, SH-3, SH-4, TRA-1-60, TRA-1-81, SSEA-4, Sox-2, and the like.
  • cell surface markers that can be used as markers of contaminating, unwanted cell types depends on the stem cell phenotype sought. For example, if collection of pluripotent hematopoietic cells is desired, contaminating cells will possess markers of commitment to the differentiated hematopoietic cells such as CD38 or CD33.
  • stem cells can be purified based on properties such as size, density, adherence to certain substrates, or ability to efflux certain dyes such as Hoechst 33342 or Rhodamine 123.
  • the stem cells can be genetically modified at any stage of the preparation. For example, a gene encoding a selectable marker or other gene of interest can be introduced to the prepared stem cells.
  • the stem cells are contacted with a composition of the invention or mixed or incubated with media that contains a composition of the invention.
  • Antibiotics, antifungals or other contamination preventive compounds can be added to the incubation media, if desired.
  • Exemplary compounds include but are not limited to penicillin, streptomycin, gentamycin, fungizone or others known in the art.
  • Stem cells in contact with compositions of the invention may be expanded through multiple passages using techniques known to those of skill in the art. For example, the stem cells isolated from a subject are counted and the viable cell density determined using trypan blue dye exclusion methods. To subculture (or passage) cells, they are replated at a density of 5,000 cells/cm 2 into a culture medium containing a composition of the invention in new tissue culture flasks coated with protein such as gelatin or fibronectin, and allowed to grow in a humidified incubator at 37° C. and 5% C0 2 . If necessary, culture medium is replenished.
  • the culture medium is removed and the adherent cells trypsinized with 0.25% trypsin and 1 mM EDTA for 3-5 min at 37°C.
  • the adherent cells are rinsed twice with PBS prior to the trypsinization.
  • the trypsin is neutralized, and the cells are pelleted by centrifugation at 300 x g for 10 min and resuspended in warm culture medium that may also contain a composition of the invention.
  • the cells are rinsed by centrifugation at 300 x g for 10 min and resuspended in warm culture medium also containing a composition of the invention.
  • Stem cells may be subcultured through multiple passages using the same passage protocol.
  • the stem cells can be contacted with compositions of the invention, for example, by simply mixing the composition(s) with the culture of stem cells. Mixing can be performed in a plethora of suitable vessels capable of maintaining viability of the stem cells. Said vessels can include but are not limited to tissue culture flasks, conical tubes, culture bags, bioreactors, or cultures that are continuously mixed. The stem cells can then be allowed to grow as desired. In some situations it will be desirable to use a combination culture system in which cells are first grown with one type of culture condition, then subsequently another culture condition is used.
  • cells when rapid expansion of hematopoietic stem cells is needed without differentiation, cells can be cultured initially in a high concentration of a composition of the invention for 48 hours, or a time period needed to induce cycling of the stem cells. Subsequently, media containing cytokines can be added into the culture for passages after the first 48 hours.
  • concentrations of the compositions of the invention can be added at different time points of the culture. For example, in a particular culture situation, addition of compositions of the invention at the initiation of culture may not be optimum.
  • the desired ratio of stem cells to compositions of the invention can be determined by one of skill in the art. For example, a ratio of less than about 1 : 1 ,000 to 1 ,000: 1 or more (stem cell preparation to composition of the invention) can be used. For example, a ratio of stem cell preparation to composition of the invention from about 1 :750, 1 :500, 1 :250, or 1 : 100 to about 100:1, 250: 1, 500: 1, or 750:1 can be used. This ratio can vary, for example, depending on temperature, incubation time, number of stem cells, the desired activity sought in the stem cells, the type of stem cells, the purity of stem cells, the amount of placental tissue used as a starting point, and the like.
  • the stem cells can be isolated from their growth media prior to contacting with a composition of the invention, or the stem cells can remain in their growth medium, with a composition of the invention added.
  • the length of time the stem cell is in contact with a composition of the invention can be determined by one of skill in the art.
  • the contacting step can range from less than about 1 second, 30 seconds, or 60 seconds to about 2 or 3 weeks or more.
  • the contacting step is between about 2, 5, 10, 30, or 45 minutes to about 12, 14, 16, 18, or 20 days. More preferably, the contacting step is between about 1, 3, 5, 8, or 24 hours to about 3, 5, 7, or 10 days.
  • conditions promoting certain types of cellular proliferation or differentiation can be used during the culture. These conditions include but are not limited to, alteration in temperature, alteration in oxygen/carbon dioxide content, alterations in turbidity of said media, or exposure to small molecules modifiers of cell cultures such as nutrients, inhibitors of certain enzymes, stimulators of certain enzymes, inhibitors of histone deacetylase activity such as valproic acid, trichostatin-A, trapoxin A, or depsipeptide, inhibitors of DNA methyltransferase activity such as 5-azacytidine, inhibitors of the enzyme GSK-3, and the like.
  • small molecules modifiers of cell cultures such as nutrients, inhibitors of certain enzymes, stimulators of certain enzymes, inhibitors of histone deacetylase activity such as valproic acid, trichostatin-A, trapoxin A, or depsipeptide, inhibitors of DNA methyltransferase activity such as 5-azacytidine, inhibitors of the enzyme GSK-3, and the like
  • stem cells such as neuronal stem cells also appear to be localized in hypoxic niches and expand preferential in low oxygen in vitro conditions as opposed to normal oxygen tension.
  • embryonic stem cells which grow at similar proliferative rates between normoxia and hypoxia, retain superior ability to form teratomas in vivo and embryoid bodies in vitro when grown under hypoxic conditions.
  • one embodiment of the invention is the use of hypoxic conditions during some or all of the incubation of the stem cells in specialized incubators with an oxygen tension ranging from 0.1% to 7.5%, preferably 0.5% to 5%, more preferably 3%-5%.
  • another embodiment of the invention is the use of hypoxic conditions in combination with compositions of the invention in order to enhance proliferation without differentiation of stem cells being grown in culture.
  • one adjuvant approach that is considered an embodiment of the invention is the use of enzymatic inhibitors in conjunction with a composition of the invention.
  • histone deacetylases are a class of enzymes involved in epigenetically opening parts of chromatin to transcription factors, thus allowing expression of genes that under normal adult conditions would not be expressed.
  • the telomerase gene catalytic subunit hTERT
  • hTERT is involved in the process of cellular immortalization and is expressed under physiological conditions only in embryonic stem cells, as well as some bone marrow hematopoietic cells, abnormally.
  • telomerase The functional role of the telomerase enzyme is to repair the shortened telomeric ends of chromosomes so that cells can escape replicative senescence.
  • Pathologically, telomerase is the enzyme responsible for the ability of cancer cells to proliferate indefinitely in cell culture. Under normal physiological conditions fibroblasts do not express telomerase and undergo replicative senescence.
  • Said acceleration was accompanied by a down-regulation of inhibitor factor p21(cip- 1/waf-l).
  • valproic acid treatment suppressed GSK3 activity and activated the Wnt signaling pathway, both of which are associated with self renewal in both hematopoietic (Gotoh, et al, 1997, Cell Growth Differ 8:721-729; Baba, et al, 2005, Immunity 23:599-609, each of which is incorporated by reference herein in its entirety), but also embryonic (Sato, et al, 2004, Nat Med 10:55-63; He, et al, 2005, Clin Lung Cancer 7:54-60, each of which is incorporated by reference herein in its entirety) stem cells.
  • valproic acid to synergize with known hematopoietic stem cell stimulatory cytokines such as Flt3L, TPO, SCF and IL-3 was demonstrated (De Felice, et al, 2005, Cancer Res 65: 1505-1513, which is incorporated by reference herein in its entirety).
  • the stem cells are able to increase their growth rate and expand rapidly.
  • culture conditions are used that allow the compositions of the invention to augment the proliferation of stem cells without induction of differentiation.
  • Any suitable method of determining the growth rate and differentiation of the stem cells can be used to determine the growth rate and cell count of the stem cells so produced. For example, flow cytometry analysis of markers associated with stem cell retention, semisolid media assays for quantification of early and committed progenitors, and in vivo NOD-SCID Repopulating Activity Assays to quantify the number of in vivo stem cells with reconstituting activity. These assays can be modified and altered in order to allow detection of specific stem cell subtypes.
  • Assays can also be developed in immune compromised mice, such as the NOD-SCID strain, by induction of a pathology to which the human stem cells is anticipated to be therapeutic.
  • human stem cells have been demonstrated to possess therapeutic activity in a variety of non- hematopoietic settings in the NOD-SCID, as well as the NUDE mouse.
  • the cell growth rate can also depend on other factors, such as, for example, temperature, type of stem cell, contents of the medium, and the time allowed for the placental incubation step and contacting step.
  • One of skill in the art will be able to alter these variables to adjust the growth rate as needed.
  • compositions, or cell culture media supplemented with the compositions of the present invention to stimulate proliferation of stem cells, including, for example:
  • human embryonic stem cells characterized by expression of markers such as SSEA-4, GCTM-2 antigen, TRA 1-60, Cripto, gastrin-releasing peptide (GRP) receptor, podocalyxin-like protein (PODXL), or human telomerase reverse transcriptase (hTERT); human oocyte producing stem cells characterized by expression of markers such Vasa, Oct-4, Dazl, Stella, Fragilis, Nobox, c-Kit and Sca-1;
  • parthenogenetically generated stem cells characterized by expression of markers such Oct-4, alkaline phosphatase, telomerase, SSEA-4, TRA 1-60 and TRA 1-81;
  • spermatogonial stem cells reprogrammed to pluripotent germ-line stem cells characterized by expression of markers such Oct-4, Nanog, Dppa5 and Rexl;
  • hematopoietic stem cells characterized by markers such as Stem Cell Antigen (SCA+), lineage negative (lin-), c-kit+, CD34+, CD38-, CD33-;
  • mesenchymal stem cells characterized by markers such as LFA-3, ICAM-1, PECAM-1, P-selectin, L-selectin, CD49b/CD29, CD49c/CD29, CD49d/CD29, CD61, CD18, CD29, 6-19, thrombomodulin, telomerase, CD10, CD13, STRO-1, STRO-2, VCAM-1, CD146, THY-1;
  • placentally derived multipotent cells characterized by markers such as Oct-4, Rex- 1, CD9, CD13, CD29, CD44, CD166, CD90, CD105, SH-3, SH-4, TRA-1-60, TRA-1-81, SSEA-4 and Sox-2;
  • adipose-derived stem cells characterized by markers such as CD 13, CD29, CD44, CD63, CD73, CD90, CD 166, Aldehyde dehydrogenase (ALDH), and ABCG2;
  • cord blood stem cells characterized by markers such as CD34, c-kit, and CXCR-4; deciduous tooth stem cells characterized by markers such as STRO-1, CD 146 (MUC18), alkaline phosphatase, MEPE, and bFGF;
  • neural stem cells characterized by markers such as RC-2, 3CB2, BLB, Sox-2hh, GLAST, Pax 6, nesting, Muashi-1, and prominin;
  • stomach epithelial stem cell characterized by markers such as Musashi-1, c-hairy-1 and HES-5;
  • skeletal muscle stem cell characterized by markers such as desmin positive, SCA- 1+, CD45- and possessing a side population profile on flow cytometry by dye exclusion; mammary gland stem cell characterized by markers such as SCA-1 positive, CD45- and keratin-6;
  • dermal stem cell characterized by markers such as SCA-1 positive, CD34+, CD45- and positive for alpha6-integrin, betal-integrin, keratin 14, and keratin 19;
  • myocardial stem cell characterized by markers such as SCA-1 positive, c-kit positive, and possessing a side population profile on flow cytometry by dye exclusion; mesangial stem cell characterized by markers such as SCA-1 positive, c-kit positive, and possessing a side population profile on flow cytometry by dye exclusion; hepatic oval stem cell characterized by markers such as SCA-1 positive, c-kit positive, and CD34 positive; or,
  • pancreatic stem cell characterized by markers such as nestin, CK-8, CK-18, Isl-1, Pdx-1, Pax-4, and Ngn-3.
  • compositions of the invention can be used as a stimulator of proliferation alone or as an additive to media known to be useful for culturing said cells.
  • tissue culture media is Dulbecco's modified Eagle's medium (DMEM).
  • compositions or cell culture media supplemented with the compositions of the present invention to stimulate proliferation of totipotent stem cells generated by cloning through the use of nuclear transfer technologies.
  • the methods of administering stem cells to a mammal comprise contacting stem cells with a composition of the invention, cultivating the stem cells under conditions suitable to facilitate the expansion of the stem cells, and introducing the expanded cells into a mammal.
  • the serum-free supplement of the present invention can also be used to prepare a stem cell type of interest for explantation into a mammal.
  • cells which have been caused to differentiate ex vivo are introduced into a mammal.
  • hematopoietic cells which have been caused to differentiate into a hematopoietic stem, precursor, or progenitor cell can be introduced into the bone marrow or the bloodstream of the mammal.
  • the differentiated cells can be introduced into, for example, the bone marrow or bloodstream of the mammal by well-known techniques.
  • a method for the expansion or growth of stem cells includes contacting a stem cell with the a growth medium containing a composition of the invention.
  • the stem cell can be
  • a totipotent cell such as an embryonic stem cell, an extra-embryonic stem cell, a cloned stem cell, a parthenogenesis derived cell
  • a pluripotent cell such as a hematopoietic stem cell, an adipose derived stem cell, a mesenchymal stem cell, a cord blood stem cell, a placentally derived stem cell, an exfoliated tooth derived stem cells, a hair follicle stem cell or a neural stem cell; or
  • tissue specific progenitor cell such as a precursor cell for the neuronal, hepatic, nephrogenic, adipogenic, osteoblastic, osteoclastic, alveolar, cardiac, intestinal, or endothelial lineage.
  • the incubating step can occur, for example, at a suitable temperature range such as from about 32°C to about 40°C.
  • the stem cells can be manipulated during their isolation or production or during their expansion or immediately prior to their administration to a mammal. Such manipulation may tailor the stem cells to perform a therapeutic function following administration to the mammal.
  • mesenchymal bone marrow cells treated with the DNA methyltransferase inhibitor 5-aza-cytidine have been shown to transdifferentiate into myocardial tissue and to improve left ventricular ejection fraction and inhibit cardiac remodeling.
  • Other types of stem cells have been utilized for improvement in myocardial activity, perfusion, and decreasing ventricular remodeling, including mesenchymal stem cells, endothelial stem cells, and skeletal myoblasts.
  • compositions and methods of this invention overcome these drawbacks by providing reproducible means of expanding and maintaining stem cell populations for subsequent therapeutic administration to a mammal.
  • DA-DKP and in particular, low molecular weight fractions of commercial human serum albumin has multiple activities that can be categorized as anti-inflammatory and remodeling/healing.
  • the anti-inflammatory properties of AmpionTM involve the inhibition of vascular permeability through cytoskeletal rearrangement in endothelial cells (cortical actin ring formation), inhibition of activation of memory T cells (not Naive) by antiCD3/CD28 or APC and reducing the amount of pro inflammatory cytokines (TNFa) produced by PBMC in response to a strong pro-inflammatory stimulus (LPS).
  • the remodeling/healing effects are through mobilization and differentiation of stem cells into tissue specific cells (such as differentiation into chondrocytes in the case of the knee).
  • TF ligand activated transcription factor
  • PPAR Peroxisome Proliferator Activator Receptor
  • RXR Retinoid X Receptor
  • PPAR belongs to the nuclear hormone receptor superfamily, expressed in inflammatory and immune cells. PPAR heterodimerizes with the retinoid X receptor (RXR) in the nucleus and together, the dimer binds to the peroxisome proliferation response element (PPRE) in the promoter of the target gene.
  • RXR Retinoid X Receptor
  • PPAR belongs to the nuclear hormone receptor superfamily, expressed in inflammatory and immune cells. PPAR heterodimerizes with the retinoid X receptor (RXR) in the nucleus and together, the dimer binds to the peroxisome proliferation response element (PPRE) in the promoter of the target gene.
  • the genes involved are anti inflammatory, involved in protection of stem cells and differentiation.
  • the N terminal A/B domain contains ligand independent activation function (AF-1), which is responsible for phosphorylation of PPAR.
  • the C domain or DNA binding domain (DBD) promotes the binding of PPAR to the PPRE in the promoter region of the target genes.
  • the D domain is the site of co-factors binding.
  • the E/F domain is the ligand binding domain (LBD) is responsible for ligand specificity and activation of PPAR binding to the PPRE which increases the expression of target genes.
  • a clinical trial was performed to investigate the effect of intra-articular knee injection of the ⁇ 5000MW Fraction ( a l s o re ferre d to h ere in a s "Amp io nTM" ) for improving joint function and reducing the pain of osteoarthritis of the knee in adults with symptomatic primary knee osteoarthritis.
  • a randomized, placebo- controlled, double-blind, parallel study with 43 evaluable subjects was chosen as the appropriate design to estimate the treatment effect and safety of the ⁇ 5000MW Fraction when it was injected into the study knee.
  • the study population was 43 patient, male or female 40-83 years old (average 63.0, standard deviation (SD) 9.6) 28 were male and 15 were female. All subjects were Caucasian. The subjects' height ranged from 162 to 192 cm (average 175.3, SD 8.1) with weight at screening ranging from 56 to 117 kg (average 88.8, SD 13.89). The subjects were fully ambulatory, with symptomatic primary knee osteoarthritis for more than 6 months prior to screening with Kellgren Lawrence Grade II or III (indicating mild or moderate osteoarthritis). Grade II for 6 subjects and Grade III for 36 subjects. One subject did have Grade IV. If both knees of a subject were osteroarthritic, one knee was selected for study while the other knee received standard of care.
  • SD standard deviation
  • the study consisted of a three week screening period and an 84 day study participation period.
  • follow-up assessments were performed at 6 hours, 24 hours and 72 hours post injection.
  • Subjects were contact by telephone at Day 8, Day 30 and Day 84 to evaluate overall pain and mobility and to monitor adverse events.
  • the subjects were offered the option of intra-articular betamethasone injection to the investigative knee for pain relief after Day 8, if deemed necessary following an assessment by the investigator.
  • the pain numerical rating scale (NRS) in the study knee was completed at pre-dose (pre-injection baseline), 6 hours post- dose on Day 1, 24 hours post-dose on Day 2, 72 hours post-dose on Day 4, and at Day 8, Day 30 and Day 84 post-dose (EOS or End-of- study).
  • the pain NRS is a numerical rating of 0-10, with 0 being no pain, 5 being moderate pain and 10 being worst possible pain.
  • the safety endpoints of the study were incidence of adverse events, vital signs at pre-dose and study Day 4, twelve lead ECG readings at screening and 24 hours post-dose, and clinical blood safety tests (biochemical and hematology) assessed at screening and 24 hours post-dose.
  • the secondary endpoints of the study were percent responders at Day 30 and Day 84, defined as an improvement in pain NRS of 2 or more points, the change from pre- injection baseline in WOMAC Osteoarthritis Index 3.1 (complete scale, pain subscore, stiffness subscore and function subscore) at 24 and 72 hours after intra-articular injection, the change from pre -injection baseline for requirement for rescue medications (paracetamol) to 24 hours and 72 hours after intra-articular injection and changes over time in mobility at Day 8, Day 30 and Day 84 post-dose compared with pre-dose and the immediate post-dose period.
  • ITT refers to subjects that met inclusion/exclusion criteria.
  • ANCOVA Analysis of covariance
  • Betamethasone injection there was no apparent difference between the use of betamethasone injections between subjects who received AmpionTM (5 of 22 subjects, 23%) compared with subjects who received saline (6 of 21 subjects, 29%).
  • Scale: -10 largest possible improvement in pain from baseline, 10 smallest possible improvement (largest increase) in pain from baseline.
  • EOS Percent responders at Day 84
  • Responder decrease in Day 84 pain NRS of -2 to -10 points (with -10 being the largest possible improvement in pain).
  • Non-responder decrease in pain at Day 84 of -1 to 10 (with 10 being the largest possible increase in pain).
  • EOS efficacy evaluable population
  • Responder decrease in Day 84 pain NRS of -2 to -10 points (with -10 being the largest possible improvement in pain).
  • Non-responder decrease in pain at Day 84 of -1 to 10 points (with 10 being the largest possible increase in pain).
  • Treatment-emergent AEs were reported for 20 of the 43 subjects (47%) following dose administration, with a total of 27 AEs. Commonly occurring AEs were headache and joint swelling and stiffness in the knee. Most subjects reported AEs classified as mild only (16 of 43 subjects, 37%). Only 4 subjects (9%) reported AEs of moderate severity:
  • Pain (as assessed by the pain numerical rating score) and WOMAC scores were reduced post-dose for each of the treatment groups for the duration of the study, except placebo at Day 84, with no significant differences between treatment groups.
  • WOMAC scores were reduced post-dose for each of the treatment groups for the duration of the study, except placebo at Day 84, with no significant differences between treatment groups.
  • Despite a higher baseline pain NRS for the saline group compared to the AmpionTM group there was a trend towards a long-term effect of study drug, with a higher percentage of subjects who responded at Day 84 for AmpionTM compared to saline.
  • In subjects receiving AmpionTM overall pain was reduced post-dose for the duration of the study, whereas subjects receiving saline did not have a reduction in pain post-dose at Day 84.
  • Use of paracetamol rescue medication up to 72 hours post-dose was highest in the Treatment E group (saline).
  • AmpionTM was considered safe and well tolerated at the dose used in the study. Improvement in pain
  • This example demonstrates the enhancement of stem cell chondrogenesis by
  • Bone marrow derived human mesenchymal stem cells Passage 5 Bone marrow derived human mesenchymal stem cells (HUXMA -01001, Cyagen Biosciences, Sunnyvale CA)
  • Cyagen Chondrogenic Differentiation medium was prepared by manufacturer protocols but excluding dexamethasone and TGF beta 3 supplements, and warmed to 37 °C in bath.
  • the treatment protocol included expanding 5 HUXMA stem cells in 182 cm 2 flasks containing 40 mis TheraPEAKTM MSCGM to 80-90% confluence. Cells were trypsinized from the flasks and a 1.0 X 10 7 cell suspension of HUXMA stem cells was warmed in Cyagen Chondrogenic Differentiation medium. 20 ⁇ of warmed cell suspension was spotted in the middle of each well of a 24-well tissue culture plate (200,000 cells per spot), and incubated at 37 °C and 5% C0 2 for one hour.
  • the plates were removed from the incubator and an additional 720 ⁇ chondrogenic medium was added to each well. 250 ⁇ saline, or drug solutions were added to the appropriate wells in triplicate (bringing the final drug dilution to one-quarter of the stock drug treatment solution concentrations). 10 ⁇ of TGF beta 3 solution (supplied by Cyagen) was added to each well, and the plates were returned to the incubator.
  • RNA isolation and analysis was conducted for all plates as follows at days 7, 14, and 22 post treatment (with the described medium exchanges). Medium from the wells was removed and saved for further protein analysis. Qiagen RNeasy plus lysis buffer (with 2mercaptoethanol) was added to each well and gently shaken for 10 minutes. The lysed cell suspensions from each well were transferred to Qiashredder columns and spun at 14,000 rpm for 2 minutes. RNA isolation proceeded by RNeasyTM plus manufacturer's protocol. RNA was eluted from the Qiagen columns using 25 ⁇ RNase free water. cDNA synthesis and real time PCR was conducted as follows. First strand synthesis of cDNA from all samples was performed in 20 ⁇ total volume using 10 ⁇ isolated RNA.
  • Collagen 2A1 Caprylate 25.88 17.67 8.21 8.477 -2.85 7.1934 7.19 6.14 1.6176
  • Collagen 2A1 Mix 28.5 16.47 12.03 10.05 0.973 0.5093 -1.96 2.00 3.4307
  • TIMP1 Dexamethasone 17.35 14.2 3.15 3.16 -0.62 1.5333 1.53 1.53 0.2057
  • TIMP1 DADKP 18.64 15.64 3 3.04 -0.77 1 .7013 1.70 1.66 0.13
  • TIMP1 Mix 19.61 16.62 2.99 3.56 -0.78 1 .7132 1.71 0.53 1.5754
  • DADKP treatment exhibited increased transcription of Collagen 2A1 and possibly Aggrecan. Visual observations suggest that the architecture of the culture may also effect the collagen transcription.
  • Example 2 Following the experiments described in Example 2, a standard procedure for testing Mesenchymal chondrogenesis protocol is developed. This examples sets forth the standardized chondrogenic testing protocol and expected results.
  • Plates were processed as follows at day 7, 14, and 22 post treatment (with the described medium exchanges).
  • MSCs bone marrow mesenchymal stem/stromal cells
  • DADKP significantly increases chondrogenesis, at least as measured by transcription of collagen 2a 1 and aggrecan.
  • NAT N-acetyl tryptophan
  • caprylate N-acetyl tryptophan
  • DADKP N-acetyl tryptophan
  • TheraPEAK MSCGM chemically defined Stem cell medium ( 190632 Lonza) • Lonza MSGM (contains serum)
  • AmpionTM expedites chondrogenesis. Also, AmpionTM has an additive or synergistic effect on the transcription or translation of genes important for the chondrocyte lineage.
  • TIMPl DADKP 18.64 15.64 3 3.04 -0.77 1.7013 1.70 1.66 0.13
  • TIMP1 Mix 19.61 16.62 2.99 3.56 -0.78 1.7132 1.71 0.53 1.5754
  • the following example is a proteomic analysis of synovial fluid from patients treated with AmpionTM compared to the synovial fluid from patients having received saline.
  • synovial fluid was taken from knees of patients in a clinical trial for use of AmpionTM to treat osteoarthritis of the knee at baseline and at 12 weeks after treatment with lOcc of either AmpionTM or saline. Baseline to 12 weeks were compared for both saline and Ampion.
  • synovial fluid samples were analyzed by SomaLogic, Inc. of Boulder,
  • the proteins shown below in Table 19 were significantly different (up or down) in the AmpionTM treated synovial fluid, compared to the saline treated synovial fluid and are known to influence cartilage and synovial fluid production.
  • Akt also known as Protein Kinase B (PKB) is a serine/threonine-specific protein kinase that plays a key role in multiple cellular processes such as cell proliferation, transcription and cell migration.
  • PKA Protein Kinase B
  • This example demonstrates the effect of intra-articular injections of a low molecular weight fraction of 5% human serum albumin (LMWF-5A) for treatment of knee pain due to osteoarthritis.
  • LMWF-5A human serum albumin
  • This study was a multicenter randomized, vehicle-controlled, double-blind, parallel study designed to evaluate the safety and efficacy of two doses of an intra-articular injection of LMWF-5A.
  • Patients with symptomatic knee osteoarthritis were randomized 1 : 1 : 1 : 1 to receive a single 4 mL or 10 mL intra-articular knee injection of either LMWF- 5 A or vehicle control (saline).
  • the primary efficacy endpoint was the difference between treatment groups in the Western Ontario and McMaster Universities (WOMAC) pain change from baseline over 12 weeks.
  • WOMAC Western Ontario and McMaster Universities
  • Safety was examined as the incidence and severity of adverse events (AEs).
  • a total of 329 patients with OA knee pain were randomized 1 : 1 : 1 : 1 across 4 study arms: 4 mL LMWF-5A, 4 mL saline vehicle control, 10 mL LMWF-5A or 10 mL saline vehicle control.
  • the patient disposition is shown in Figure 10.
  • the starting material of LMWF-5A HSA purchased from OctaPharma (Lachen, Switzerland), was subjected to centrifugation/ultrafiltration under sterile conditions and the ultrafiltrate, containing species with a MW less than 5000 Da, was separated.
  • the ultrafiltrate contained DA-DKP (approximately 50 - 200 mM) and the excipients (i.e. sodium caprylate and sodiumacetyltryptophanate).
  • the ultrafiltrate was transferred for aseptic filling, to afford sterile drug product.
  • osteoarthritis Index 3.1 5-point Likert score the Patient's Global Assessment of disease severity (PGA) using a 5-point Likert Score, and the amount of acetaminophen after intraarticular injection.
  • Acetaminophen was supplied in 500 mg tablets at baseline as a rescue medication, and allowed as 1 tablet every 4 hours as needed.
  • Safety was evaluated by recording adverse events (through 24 hours post-dose and at all follow-up contacts), vital signs and physical examination results (baseline, weeks 6 and 12).
  • Example 8 This example is a twenty week extension of the clinical trial described in Example 8 demonstrating the effects of LMWF-5 A for treatment of knee pain due to osteoarthritis.
  • NMT01839331 This analysis is a 20-week extension of a multicenter, randomized, vehicle- controlled, double-blind study (NCT01839331) that evaluated the efficacy and safety of the low molecular weight fraction of 5% human serum albumin (LMWF-5 A) for treatment of inflammation-associated pain in symptomatic OA of the knee (OAK).
  • LMWF-5 A human serum albumin

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Pain & Pain Management (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/US2014/030538 2013-03-15 2014-03-17 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same WO2014145729A2 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
CN201480015832.9A CN105101965B (zh) 2013-03-15 2014-03-17 用于干细胞活动化、归巢、扩增和分化的组合物及其使用方法
SG11201506267XA SG11201506267XA (en) 2013-03-15 2014-03-17 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
CA2906864A CA2906864A1 (en) 2013-03-15 2014-03-17 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
EP14763038.8A EP2968315B1 (en) 2013-03-15 2014-03-17 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
AU2014232728A AU2014232728B2 (en) 2013-03-15 2014-03-17 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
MX2015010937A MX2015010937A (es) 2013-03-15 2014-03-17 Composiciones para la movilizacion, autodireccion, expansion y diferenciacion de celulas madre y metodos para usar las mismas.
EA201500943A EA201500943A1 (ru) 2013-03-15 2014-03-17 Составы для мобилизации, хоуминга, размножения и дифференцировки стволовых клеток и способы применения составов
JP2016503416A JP6588005B2 (ja) 2013-03-15 2014-03-17 軟骨形成を刺激するために使用される医薬組成物
KR1020157029761A KR20150132508A (ko) 2013-03-15 2014-03-17 줄기세포의 가동화, 회귀, 증식 및 분화를 위한 조성물 및 이의 사용 방법
BR112015020469A BR112015020469A2 (pt) 2013-03-15 2014-03-17 Usos de da-dkp, composição e método para fornecimento de células-tronco a umindivíduo
NZ712630A NZ712630A (en) 2013-03-15 2014-03-17 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
IL240473A IL240473A0 (en) 2013-03-15 2015-08-10 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods for their use
PH12015502048A PH12015502048A1 (en) 2013-03-15 2015-09-10 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
ZA2015/07291A ZA201507291B (en) 2013-03-15 2015-10-01 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
HK16102085.4A HK1214143A1 (zh) 2013-03-15 2016-02-24 用於干細胞活動化、歸巢、擴增和分化的組合物及其使用方法

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US201361791623P 2013-03-15 2013-03-15
US61/791,623 2013-03-15
US201361832713P 2013-06-07 2013-06-07
US61/832,713 2013-06-07
US201361897449P 2013-10-30 2013-10-30
US61/897,449 2013-10-30
US201461923314P 2014-01-03 2014-01-03
US61/923,314 2014-01-03
US201461939625P 2014-02-13 2014-02-13
US61/939,625 2014-02-13

Publications (2)

Publication Number Publication Date
WO2014145729A2 true WO2014145729A2 (en) 2014-09-18
WO2014145729A3 WO2014145729A3 (en) 2014-12-24

Family

ID=51538532

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/030538 WO2014145729A2 (en) 2013-03-15 2014-03-17 Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same

Country Status (17)

Country Link
US (3) US9808454B2 (ru)
EP (1) EP2968315B1 (ru)
JP (1) JP6588005B2 (ru)
KR (1) KR20150132508A (ru)
CN (1) CN105101965B (ru)
AU (1) AU2014232728B2 (ru)
BR (1) BR112015020469A2 (ru)
CA (1) CA2906864A1 (ru)
EA (1) EA201500943A1 (ru)
HK (1) HK1214143A1 (ru)
IL (1) IL240473A0 (ru)
MX (1) MX2015010937A (ru)
NZ (1) NZ712630A (ru)
PH (1) PH12015502048A1 (ru)
SG (2) SG11201506267XA (ru)
WO (1) WO2014145729A2 (ru)
ZA (1) ZA201507291B (ru)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3183240A4 (en) * 2014-08-18 2018-06-27 Ampio Pharmaceuticals, Inc. Treatment of joint conditions

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2417960C (en) 2000-08-04 2012-07-10 Dmi Biosciences, Inc. Method of using diketopiperazines and composition containing them
CN104095851A (zh) 2003-05-15 2014-10-15 安皮奥制药股份有限公司 T-细胞介导的疾病的治疗
EP2300011A4 (en) 2008-05-27 2012-06-20 Dmi Life Sciences Inc METHODS AND THERAPEUTIC COMPOUNDS
US8507496B2 (en) 2010-09-07 2013-08-13 Dmi Acquisition Corp. Treatment of diseases
KR20140075772A (ko) 2011-10-10 2014-06-19 앰피오 파마슈티컬스 인코퍼레이티드 면역 내성을 증가시킨 이식 의료 장비, 및 그의 제조 및 이식 방법
PL2766029T3 (pl) 2011-10-10 2020-08-24 Ampio Pharmaceuticals, Inc. Leczenie choroby zwyrodnieniowej stawów
CN103841974A (zh) 2011-10-28 2014-06-04 安皮奥制药股份有限公司 鼻炎的治疗
MX2015010937A (es) 2013-03-15 2015-10-29 Ampio Pharmaceuticals Inc Composiciones para la movilizacion, autodireccion, expansion y diferenciacion de celulas madre y metodos para usar las mismas.
EP3310375A4 (en) 2015-06-22 2019-02-20 Ampio Pharmaceuticals, Inc. USE OF LOW MOLECULAR WEIGHT HUMAN SERUM ALBUMIN FRACTIONS FOR TREATING DISEASES
EP3461887A4 (en) 2016-03-09 2020-01-22 University Of Ulsan Foundation For Industry Cooperation COMPOSITION FOR PROLIFERATION, PROMOTION OF DIFFERENTIATION OR SENESCENCE INHIBITION OF STEM CELLS WITH JAK1 INHIBITOR AS AN ACTIVE SUBSTANCE
WO2017160986A1 (en) * 2016-03-16 2017-09-21 Cell Medicine, Inc. Mesenchymal stem cells with enhanced efficacy
TWI744300B (zh) * 2016-03-23 2021-11-01 里爾中央醫學中心 改良性熱處理的血小板顆粒裂解液在製備用於治療神經系統疾病的組合物的用途
CN107418934B (zh) * 2016-05-24 2021-01-01 南开大学 Tnfsf15蛋白在人脐带血造血干细胞体外扩增中的用途
US10456423B2 (en) 2016-06-13 2019-10-29 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10426796B2 (en) * 2016-06-13 2019-10-01 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
CN106834223B (zh) * 2017-04-05 2020-03-27 上海逍鹏生物科技有限公司 诱导脐带间充质干细胞向软骨细胞分化的方法
CN112094804B (zh) * 2019-06-18 2024-05-14 中国医学科学院基础医学研究所 一种异质性干细胞群、其制备方法及用途
CN111073857A (zh) * 2019-12-30 2020-04-28 深圳爱生再生医学科技有限公司 一种治疗关节炎的干细胞生物制品及其制备方法与应用
KR102545746B1 (ko) * 2020-12-03 2023-06-20 가톨릭대학교 산학협력단 유도만능줄기세포 유래 혈관세포 및 SDF1a를 발현하는 중간엽 줄기세포를 포함하는 심장 재생용 조성물
CN112657048B (zh) * 2021-01-26 2024-04-02 南方科技大学 一种非手性磁控微型机器人及其制备方法和应用
CN114209814B (zh) * 2021-12-15 2024-02-23 南开大学 Tnfsf15蛋白在促进骨髓干细胞分化为巨噬细胞并扩增中的用途
CN114717184B (zh) * 2022-05-05 2023-04-21 中国水产科学研究院北戴河中心实验站 一种牙鲆精原干细胞培养液及建立牙鲆精原干细胞系的方法

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4694081A (en) 1985-09-23 1987-09-15 Monsanto Company Process to prepare 2,5-diketopiperazines
WO1996000391A1 (en) 1994-06-23 1996-01-04 Affymax Technologies N.V. Methods for the synthesis of diketopiperazines
WO1997048685A1 (en) 1996-06-18 1997-12-24 Glaxo Group Limited Metalloprotease inhibitors
US5817751A (en) 1994-06-23 1998-10-06 Affymax Technologies N.V. Method for synthesis of diketopiperazine and diketomorpholine derivatives
US5932579A (en) 1996-06-18 1999-08-03 Affymax Technologies N.V. Collagenase-1 and stromelysin-1 inhibitors, pharmaceutical compositions comprising same and methods of their use
US6555543B2 (en) 2000-08-04 2003-04-29 Dmi Biosciences, Inc. Method of using diketopiperazines and composition containing them
US20040024180A1 (en) 2000-04-20 2004-02-05 Karlheinz Drauz Process for the production of 2,5 -diketopiperazines,2,5-diketopiperazines , dipeptides and their use thereof

Family Cites Families (208)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3941790A (en) 1967-07-03 1976-03-02 National Research Development Corporation Bis diketopiperazines
BE765030A (fr) 1970-04-02 1971-08-16 Snam Progetti Composes contenant un noyau de dicetopiperazine et leur procedede fabrication
GB1353304A (en) 1971-03-30 1974-05-15 Sagami Chem Res Process for production of diketopiperazine dihydroxamates and intermediate therefor
US4006261A (en) 1973-09-28 1977-02-01 Firmenich S.A. Flavoring with mixtures of theobromine and cyclic dipeptides
US3928330A (en) 1973-12-19 1975-12-23 Ciba Geigy Corp Substituted piperazinedione carboxylic acids and metal salts thereof
GB1459488A (en) 1974-03-19 1976-12-22 Wyeth John & Brother Ltd Piperazinedione derivatives
US4088649A (en) 1975-07-02 1978-05-09 Firmenich S.A. Process for preparing a diketo-piperazine
JPS5225019A (en) 1975-08-19 1977-02-24 New Zealand Inventions Dev Production of serum albumin
US4205057A (en) 1978-01-17 1980-05-27 Government Of The United States Cerebrospinal fluid protein fragments
DE2940654A1 (de) 1979-10-06 1981-04-16 Bayer Ag, 5090 Leverkusen Dimeres keten der 1,2,,-triazol-3-carbonsaeure
US4289759A (en) 1980-06-23 1981-09-15 Ortho Pharmaceutical Corporation Immunoregulatory diketopiperazine compounds
US4331595A (en) 1980-06-23 1982-05-25 Ortho Pharmaceutical Corporation Immunoregulatory diketopiperazine compounds
JPS5836225U (ja) 1981-08-31 1983-03-09 株式会社東芝 金属製平パレツト
JPS5973574A (ja) 1982-10-21 1984-04-25 Grelan Pharmaceut Co Ltd 環状ジペプチド類
US4694061A (en) 1983-10-12 1987-09-15 Ciba-Geigy Corporation Radiation-sensitive polycondensates, processes for their preparation coated material and its use
US4727018A (en) 1984-05-18 1988-02-23 Eichner Ronald D Immunoregulation of transplantable tissue
JPS61112060A (ja) 1984-11-02 1986-05-30 Fujisawa Pharmaceut Co Ltd ピペラジン化合物
US4806538A (en) 1984-11-02 1989-02-21 Fujisawa Pharmaceutical Co., Ltd. Piperazine compound as PAF-antagonist
CS254868B1 (cs) 1985-06-11 1988-02-15 Jiri Vanzura Způsob výroby cyklických dipeptidů
JPS6236331A (ja) 1985-08-12 1987-02-17 Kenji Suzuki 抗感染症剤ならびに免疫賦活剤
US4771056A (en) 1985-08-29 1988-09-13 Roman Rozencwaig Method of medical treatment with serotonin antagonists
US4661500A (en) 1985-08-29 1987-04-28 Roman Rozencwaig Method of medical treatment with serotonin antagonists
US5047401A (en) 1985-09-09 1991-09-10 Board Of Regents, The University Of Texas System Use of dipeptide alkyl esters to treat GVHD
PT83613B (en) 1985-10-28 1988-11-21 Lilly Co Eli Process for the selective chemical removal of a protein amino-terminal residue
JPS63290868A (ja) 1987-05-22 1988-11-28 Fujisawa Pharmaceut Co Ltd ジケトピペラジン誘導体およびその塩類
JPS6413075A (en) 1987-07-03 1989-01-17 Nippon Chemiphar Co Production of 2, 5-piperazinedione derivative
US5512544A (en) 1987-09-13 1996-04-30 Yeda Research And Development Co. Ltd. Pharmaceutical compositions comprising an anticytokine
US4992552A (en) 1988-08-31 1991-02-12 Eastman Kodak Company Process for preparation of amino acids
US5144073A (en) 1988-08-31 1992-09-01 Hubbs John C Process for preparation of dipeptides
JP2001055340A (ja) 1988-10-31 2001-02-27 Welfide Corp アルブミン製剤の製造方法
US5238938A (en) 1989-02-10 1993-08-24 Otsuka Pharmaceutical Co., Ltd. Indole derivatives
DE3906952A1 (de) 1989-03-04 1990-09-06 Boehringer Mannheim Gmbh (3-(c(pfeil abwaerts)1(pfeil abwaerts)(pfeil abwaerts)6(pfeil abwaerts)-c(pfeil abwaerts)1(pfeil abwaerts)(pfeil abwaerts)8(pfeil abwaerts))alkansulfinyl- und sulfonyl-2-methoxymethyl-propyl)-(2-trimethylammonio-ethyl) phosphate, verfahren zu deren herstellung diese verbindungen enthaltende arzneimittel
US5902790A (en) 1995-10-03 1999-05-11 Cytran, Inc. Pharmaceutical angiostatic dipeptide compositions and method of use thereof
ES2070878T3 (es) 1989-10-13 1995-06-16 Phobos Nv Procedimiento para la produccion en continuo de resina de poliester de alto peso molecular.
JPH03176478A (ja) 1989-12-05 1991-07-31 Snow Brand Milk Prod Co Ltd 新規なジケトピペラジン誘導体及びこれを有効成分とする免疫抑制剤
EP0478729B1 (en) 1990-03-15 1996-08-21 The Nutrasweet Company Process for manufacturing aspartame from a diketopiperazine and novel intermediates and derivatives therefor
US6180616B1 (en) 1990-05-10 2001-01-30 Atsuo F. Fukunaga Use of purine receptor agonists to alleviate or normalize physiopathologically excited sensory nerve function
US6099856A (en) 1992-06-15 2000-08-08 Emisphere Technologies, Inc. Active agent transport systems
US5578323A (en) 1992-06-15 1996-11-26 Emisphere Technologies, Inc. Proteinoid carriers and methods for preparation and use thereof
US5693338A (en) 1994-09-29 1997-12-02 Emisphere Technologies, Inc. Diketopiperazine-based delivery systems
US6331318B1 (en) 1994-09-30 2001-12-18 Emisphere Technologies Inc. Carbon-substituted diketopiperazine delivery systems
GB9022543D0 (en) 1990-10-17 1990-11-28 Wellcome Found Antibody production
JP3266612B2 (ja) 1990-11-14 2002-03-18 ザ ユニバーシティー カンパニー プロプライエタリーリミテッド 多発性硬化症の診断および治療
US5776892A (en) 1990-12-21 1998-07-07 Curative Health Services, Inc. Anti-inflammatory peptides
JPH04234374A (ja) 1990-12-27 1992-08-24 Ajinomoto Co Inc ジケトピペラジン誘導体の製造方法
GB9102997D0 (en) 1991-02-13 1991-03-27 Pfizer Ltd Therapeutic agents
US5543503A (en) 1991-03-29 1996-08-06 Genentech Inc. Antibodies to human IL-8 type A receptor
US5538993A (en) 1991-09-12 1996-07-23 Yissum Research Development Company Certain tetrahydrocannabinol-7-oic acid derivatives
AU2905092A (en) 1991-10-28 1993-06-07 Cytoven Pharmaceutical dipeptide compositions and methods of use thereof
JPH05244982A (ja) 1991-12-06 1993-09-24 Sumitomo Chem Co Ltd 擬人化b−b10
JPH05163148A (ja) 1991-12-18 1993-06-29 Kanebo Ltd 抗腫瘍剤
GB9200210D0 (en) 1992-01-07 1992-02-26 British Bio Technology Compounds
US5352461A (en) 1992-03-11 1994-10-04 Pharmaceutical Discovery Corporation Self assembling diketopiperazine drug delivery system
US5418218A (en) 1992-07-10 1995-05-23 The University Of Maryland At Baltimore Histidyl-proline diketopiperazine (cyclo his-pro) a cns-active pharmacologic agent
US5463083A (en) 1992-07-13 1995-10-31 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5648486A (en) 1992-07-13 1997-07-15 Cytomed, Inc. Compounds and methods for the treatment of inflammatory and immune disorders
US5358938A (en) 1992-07-13 1994-10-25 Cytomed, Inc. Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase
US5434151A (en) 1992-08-24 1995-07-18 Cytomed, Inc. Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase
GB9217331D0 (en) 1992-08-14 1992-09-30 Xenova Ltd Pharmaceutical compounds
EP0656004A1 (en) 1992-08-20 1995-06-07 Cytomed, Inc. Dual functional anti-inflammatory and immunosuppressive agents
US5728553A (en) 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
PL177771B1 (pl) 1992-12-18 2000-01-31 Cell Med Inc Linie komórek hybrydoma i preparaty przeciwciał
ES2068742B1 (es) 1993-02-11 1995-11-16 Uriach & Cia Sa J Nuevos derivados de piridinio.
WO1994020063A2 (en) 1993-03-04 1994-09-15 Cytoven International N.V. Pharmaceutical tryptophan containing dipeptide compositions and methods of use thereof
US6107050A (en) 1993-05-03 2000-08-22 The United States Of America As Represented By The Department Of Health And Human Services Diagnostic test for alzheimers disease
US5523226A (en) * 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
WO1995003054A1 (en) 1993-07-23 1995-02-02 Lxr Biotechnology Inc. Methods of treating apoptosis and associated conditions
GB9324872D0 (en) 1993-12-03 1994-01-19 Univ Pasteur Pharmaceutical compounds
IL112627A0 (en) 1994-02-14 1995-05-26 Xenova Ltd Diketopiperazines, their preparation and pharmaceutical or veterinary compositions containing them
JP2921731B2 (ja) 1994-03-10 1999-07-19 日清製油株式会社 有機溶媒または油脂類のゲル化剤
FR2717484A1 (fr) 1994-03-16 1995-09-22 Pf Medicament Nouveaux composés pseudo-bis-peptidiques analogues de la CCK, leur procédé de préparation, leur utilisation à titre de médicament et les compositions pharmaceutiques les comprenant .
GB9410387D0 (en) 1994-05-24 1994-07-13 Xenova Ltd Pharmaceutical compounds
US5550132A (en) 1994-06-22 1996-08-27 University Of North Carolina Hydroxyalkylammonium-pyrimidines or purines and nucleoside derivatives, useful as inhibitors of inflammatory cytokines
PL317873A1 (en) 1994-06-27 1997-04-28 Cytomed Inc Compounds for and method of treating cardiovascular, inflammatory and immunological disorders
US5703093A (en) 1995-05-31 1997-12-30 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5792776A (en) 1994-06-27 1998-08-11 Cytomed, Inc., Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5750565A (en) 1995-05-25 1998-05-12 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5843950A (en) 1994-07-29 1998-12-01 Nikken Chemicals Co., Ltd. 1,4-dihydropyridine compound and pharmaceutical composition containing the same
US5561115A (en) 1994-08-10 1996-10-01 Bayer Corporation Low temperature albumin fractionation using sodium caprylate as a partitioning agent
WO1996011009A1 (en) 1994-10-05 1996-04-18 Cari Loder Treatment of multiple sclerosis (ms) and other demyelinating conditions using lofepramine in combination with l-phenylalanine, tyrosine or tryptophan and possibly a vitamin b12 compound
ES2087038B1 (es) 1994-11-07 1997-03-16 Uriach & Cia Sa J Nuevas piperidinas con actividad antagonista del paf.
CZ282794A3 (en) 1994-11-16 1996-04-17 Galena Pentapeptidic precursors of biologically active cyclic dipeptides
EP0805149A4 (en) 1994-12-01 1998-03-11 Toyama Chemical Co Ltd NEW 2,3-DICETOPIPERAZINE DERIVATIVE OR SALT THEREOF
JP3634891B2 (ja) 1995-04-06 2005-03-30 株式会社海洋バイオテクノロジー研究所 キチナーゼ阻害物質
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
AU6311996A (en) 1995-07-06 1997-02-05 Zeneca Limited Peptide inhibitors of fibronectine
US5811241A (en) 1995-09-13 1998-09-22 Cortech, Inc. Method for preparing and identifying N-substitued 1,4-piperazines and N-substituted 1,4-piperazinediones
US5665714A (en) 1995-12-07 1997-09-09 Clarion Pharmaceuticals Inc. N-substituted glycerophosphoethanolamines
US6060452A (en) 1996-03-13 2000-05-09 Cytran, Inc. Analogs of L-Glu-L-Trp having pharmacological activity
US6541224B2 (en) 1996-03-14 2003-04-01 Human Genome Sciences, Inc. Tumor necrosis factor delta polypeptides
EP0891349A4 (en) 1996-04-03 2001-01-24 Merck & Co Inc FARNESYL-PROTEIN TRANSFERASE INHIBITORS
US5919785A (en) 1996-04-03 1999-07-06 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
HUP9901254A3 (en) 1996-04-03 2001-10-29 Perpres As Non-dendritic backbone peptide carrier
WO1997038688A1 (en) 1996-04-12 1997-10-23 Peptide Technology Pty. Limited Methods of treating immunopathologies using polyunsaturated fattyacids
US5985581A (en) 1996-07-25 1999-11-16 The Mclean Hospital Corporation Use of presenilin-1 for diagnosis of alzheimers disease
WO1998009968A1 (en) 1996-09-09 1998-03-12 Novartis Animal Health Australasia Pty. Limited Terpenylated diketopiperazines, (drimentines)
NZ335544A (en) 1996-10-04 2001-08-31 Neuronz Ltd Use of GPE (tripeptide or dipeptide) in form of Gly-Pro-Glu or Gly-Pro or Pro-Glu as a neuromodulator
RU2125728C1 (ru) 1996-10-10 1999-01-27 Пермская государственная медицинская академия Способ иммунодиагностики рассеянного склероза
EP0835660A1 (en) 1996-10-14 1998-04-15 Gaston Edmond Filomena Merckx Products containing methylcobalamin for the treatment of multiple sclerosis or other demyelinating conditions
US6441172B1 (en) 1996-11-07 2002-08-27 Torrey Pines Institute For Molecular Studies Diketodiazacyclic compounds, diazacyclic compounds and combinatorial libraries thereof
US5932112A (en) 1996-11-27 1999-08-03 Browning Transport Management, Inc. Method and apparatus for killing microorganisms in ship ballast water
RU2112242C1 (ru) 1996-12-09 1998-05-27 Пермская государственная медицинская академия Способ диагностики рассеянного склероза
JPH10226615A (ja) 1997-02-18 1998-08-25 Pola Chem Ind Inc アスパラギン酸フェニルアラニン環状ジペプタイド誘導体を含有する組成物
JPH10245315A (ja) 1997-03-05 1998-09-14 Pola Chem Ind Inc シクロジペプタイド誘導体を含有する組成物
US6306909B1 (en) 1997-03-12 2001-10-23 Queen's University At Kingston Anti-epileptogenic agents
AU6465798A (en) 1997-03-14 1998-09-29 Neuromark Diagnosing neurologic disorders
US6265535B1 (en) 1997-05-30 2001-07-24 The Trustees Of The University Of Pennsylvania Peptides and peptide analogues designed from binding sites of tumor necrosis factor receptor superfamily and their uses
RU2128840C1 (ru) 1997-06-16 1999-04-10 Пермская государственная медицинская академия Способ диагностики рассеянного склероза
US6222029B1 (en) 1997-08-01 2001-04-24 Genset 5′ ESTs for secreted proteins expressed in brain
US5834032A (en) 1997-08-11 1998-11-10 Song; Moon K. Compositions and methods for treating diabetes
US7202279B1 (en) 1998-02-11 2007-04-10 Georgetown University Cyclic dipeptides and azetidinone compounds and their use in treating CNS injury and neurodegenerative disorders
EP0939124A3 (en) 1998-02-24 2001-03-21 Smithkline Beecham Plc MGBP1 sequences
WO1999049865A1 (en) 1998-03-31 1999-10-07 Mayo Foundation For Medical Education And Research Use of platelet activating factor (paf) inhibitors to inhibit il-5 induced eosinophil activation or degranulation
EP1067951A2 (en) 1998-04-03 2001-01-17 Cytran Ltd. Use of l-glu-l-trp in the treatment of hiv infection
WO1999051720A2 (en) 1998-04-03 1999-10-14 Cytran Ltd. Methods for production of therapeutic cytokines
AU767241B2 (en) 1998-09-14 2003-11-06 Qiang Xu Immunosuppressive agents
US6475743B1 (en) 1998-10-02 2002-11-05 Ischemia Technologies, Inc. Marker useful for detection and measurement of free radical damage and method
WO2000020840A1 (en) 1998-10-02 2000-04-13 Ischemia Technologies, Inc. Tests for the rapid evaluation of ischemic states and kits
US6461875B1 (en) 1998-10-02 2002-10-08 Ischemia Technologies, Inc. Test for rapid evaluation of ischemic states and kit
DE69939121D1 (de) 1998-10-02 2008-08-28 Ischemia Tech Inc Verfahren und materialien zum nachweis und zur messung von durch freie radikale verursachten schäden
US6492179B1 (en) 1998-10-02 2002-12-10 Ischemia Techologies, Inc. Test for rapid evaluation of ischemic states and kit
DE19847690A1 (de) 1998-10-15 2000-04-20 Brahms Diagnostica Gmbh Verfahren und Substanzen für die Diagnose und Therapie von Sepsis und sepsisähnlichen systemischen Infektionen
US7026322B2 (en) 1998-11-12 2006-04-11 Nereus Pharmaceuticals, Inc. Phenylahistin and the phenylahistin analogs, a new class of anti-tumor compounds
US6358957B1 (en) 1998-11-12 2002-03-19 Nereus Pharmaceuticals, Inc. Phenylahistin and the phenylahistin analogs, a new class of anti-tumor compounds
WO2000043000A1 (fr) 1999-01-20 2000-07-27 Kyowa Hakko Kogyo Co., Ltd. Inhibiteurs de proteasome
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
WO2000057187A2 (en) 1999-03-19 2000-09-28 Vanderbilt University Diagnosis and treatment of multiple sclerosis
US6090780A (en) 1999-04-07 2000-07-18 Chandon Prasad Histidyl-proline diketopiperazine and method of use
US6689765B2 (en) 1999-05-04 2004-02-10 Schering Corporation Piperazine derivatives useful as CCR5 antagonists
JP2000327575A (ja) 1999-05-26 2000-11-28 Teika Seiyaku Kk ジケトピペラジン誘導体含有炎症疾患治療剤および新規なジケトピペラジン誘導体
DE19937721A1 (de) 1999-08-10 2001-02-15 Max Planck Gesellschaft Neue Diketopiperazine
CN1507437A (zh) 1999-11-12 2004-06-23 具有5-羟色胺5-ht1a活性的支链金刚烷基及降金刚烷基芳基-与芳烷基-哌嗪
US6677473B1 (en) 1999-11-19 2004-01-13 Corvas International Inc Plasminogen activator inhibitor antagonists
US20050096323A1 (en) 1999-11-30 2005-05-05 Novoscience Pharma Inc. Diketopiperazine derivatives to inhibit thrombin
JP4683810B2 (ja) 2000-02-29 2011-05-18 中外製薬株式会社 長期安定化製剤
US7288545B2 (en) 2000-05-09 2007-10-30 Angiorx Corporation Piperazinedione compounds
DE60137037D1 (de) 2000-05-09 2009-01-29 Angiorx Corp Piperazindion-verbindungen
DE10026998A1 (de) 2000-05-31 2001-12-13 Fresenius Kabi De Gmbh Verfahren zur Herstellung einer kosmetischen Zusammensetzung, die humanes Serum Albumin umfasst, welches aus transgenen nicht-menschlichen Säugern erhalten wurde
JP2004533402A (ja) 2000-06-08 2004-11-04 リリー アイコス リミテッド ライアビリティ カンパニー Pdevインヒビターとしての四環系ジケトピペラジン化合物
EP1311277A4 (en) 2000-07-18 2004-08-25 Joslin Diabetes Center Inc FIBROSIS MODULATION METHOD
CZ20002681A3 (cs) 2000-07-21 2002-03-13 Ev®En Ing. Csc. Kasafírek Cyklické tyrosinové dipeptidy
CZ20002680A3 (cs) 2000-07-21 2002-03-13 Ev®En Ing. Csc. Kasafírek Cyklické alkylthiopeptidy
US6967202B2 (en) 2000-08-04 2005-11-22 Dmi Biosciences, Inc. Method of synthesizing diketopiperazines
US20070208087A1 (en) 2001-11-02 2007-09-06 Sanders Virginia J Compounds, compositions and methods for the treatment of inflammatory diseases
AU2001297611A1 (en) 2000-12-29 2002-08-19 Celltech R And D, Inc. Pharmaceutical uses and synthesis of diketopiperazines
GB2372740A (en) 2001-01-17 2002-09-04 Xenova Ltd Diketopiperazines
WO2002059604A2 (en) 2001-01-26 2002-08-01 Oxford Glycosciences (Uk) Ltd Diagnosis and treatment of multiple sclerosis
JP2004535381A (ja) 2001-04-13 2004-11-25 バーテックス ファーマシューティカルズ インコーポレイテッド c−JunN末端キナーゼ(JNK)および他のプロテインキナーゼのインヒビター
DE60229059D1 (de) 2001-05-08 2008-11-06 Univ Yale Proteomimetische verbindungen und verfahren
US7368421B2 (en) 2001-06-27 2008-05-06 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitors in the treatment of multiple sclerosis
WO2003032809A2 (en) 2001-10-15 2003-04-24 The Medstar Research Institute Modulation of akt-dependent response to prevent restenosis
EP1575976A4 (en) 2001-11-02 2006-08-23 Insert Therapeutics Inc METHODS AND COMPOSITIONS FOR THE THERAPEUTIC USE OF RNA INTERFERENCE
US20040063654A1 (en) 2001-11-02 2004-04-01 Davis Mark E. Methods and compositions for therapeutic use of RNA interference
GB0128108D0 (en) 2001-11-23 2002-01-16 Astrazeneca Ab Therapeutic use
CN101671707A (zh) 2001-12-27 2010-03-17 味之素株式会社 谷氨酸衍生物的制备方法
MXPA04006928A (es) 2002-01-18 2004-12-06 Unilever Nv Composiciones cosmeticas comprendiendo un compuesto de ciclodipeptido.
CA2480832A1 (en) 2002-04-05 2003-10-23 Nitromed, Inc. Nitric oxide donors, compositions and methods of use
ES2204294B2 (es) 2002-07-02 2005-02-01 Universidade De Santiago De Compostela Nuevos antibioticos activos frente al vibrio anguillarum y sus aplicaciones en cultivos de peces, crustaceos, moluscos y otras actividades de acuicultura.
US20040038865A1 (en) 2002-08-01 2004-02-26 Mannkind Corporation Cell transport compositions and uses thereof
US20080260838A1 (en) 2003-08-01 2008-10-23 Mannkind Corporation Glucagon-like peptide 1 (glp-1) pharmaceutical formulations
US7919497B2 (en) 2002-08-02 2011-04-05 Nereus Pharmaceuticals, Inc. Analogs of dehydrophenylahistins and their therapeutic use
CA2494049C (en) 2002-08-02 2011-10-18 Nereus Pharmaceuticals, Inc. Dehydrophenylahistins and analogs thereof and the synthesis of dehydrophenylahistins and analogs thereof
KR20050042146A (ko) 2002-08-06 2005-05-04 아플라겐 게엠베하 결합 분자
DE10238144A1 (de) 2002-08-15 2004-02-26 Basf Ag Verwendung von Diketopiperazin-Derivaten als photostabile UV-Filter in kosmetischen und pharmazeutischen Zubereitungen
WO2004022569A1 (en) 2002-09-03 2004-03-18 Georgetown University Akt inhibitors, pharmaceutical compositions, and uses thereof
DK1571970T3 (da) 2002-10-02 2011-11-28 Dmi Biosciences Inc Diagnose og monitorering af sygdomme
US7196169B2 (en) 2002-10-11 2007-03-27 Queen's University At Kingston Isolated post-translationally modified mammalian proteins for monitoring and diagnosing muscle damage
EP1572669A2 (en) 2002-11-22 2005-09-14 Novo Nordisk A/S 2,5-diketopiperazines for the treatment of obesity
CN104095851A (zh) 2003-05-15 2014-10-15 安皮奥制药股份有限公司 T-细胞介导的疾病的治疗
CN1791420A (zh) * 2003-05-15 2006-06-21 Dmi生物科学公司 T-细胞介导的疾病的治疗
PT1651620E (pt) 2003-07-30 2012-02-10 Xenon Pharmaceuticals Inc Derivados de piperizina e sua utilização como agentes terapêuticos
EP1664050B1 (en) 2003-09-03 2015-12-09 Neuren Pharmaceuticals Limited Neuroprotective bicyclic compounds and pharmaceutical compositions containing them
EP1768986A2 (en) 2004-06-29 2007-04-04 Amgen Inc. Furanopyrimidines
WO2006023831A2 (en) 2004-08-20 2006-03-02 Prometic Biosciences Ltd. Sequential protein isolation and purification schemes by affinity chromatography
KR20130066695A (ko) 2004-08-23 2013-06-20 맨카인드 코포레이션 약물 전달용 디케토피페라진염, 디케토모르포린염 또는 디케토디옥산염
JP2008530111A (ja) 2005-02-14 2008-08-07 メルク エンド カムパニー インコーポレーテッド Akt活性の阻害剤
US20070065483A1 (en) 2005-09-21 2007-03-22 Chudzik Stephen J In vivo formed matrices including natural biodegradable polysaccharides and uses thereof
MX2008010721A (es) 2006-02-22 2008-09-01 Mannkind Corp Un metodo para mejorar las propiedades farmaceuticas de microparticulas que contienen dicetopiperazina y un agente activo.
MX2008013216A (es) 2006-04-14 2008-10-27 Mannkind Corp Formulaciones farmaceuticas de peptido-1 tipo glucagon (glp-1).
US20080017576A1 (en) 2006-06-15 2008-01-24 Rensselaer Polytechnic Institute Global model for optimizing crossflow microfiltration and ultrafiltration processes
AR061240A1 (es) 2006-06-20 2008-08-13 Lilly Co Eli Compuestos de de 4-isoquinolin-fenol,composiciones farmaceuticas que los contienen y su uso como agentes antineoplasicos y/o antivirales.
AU2007272954A1 (en) 2006-07-11 2008-01-17 Harkness Pharmaceuticals, Inc. Methods of treating obesity using satiety factors
US8231929B2 (en) 2006-11-09 2012-07-31 Cook Medical Technologies Llc Medical device coating process
WO2009009793A2 (en) 2007-07-12 2009-01-15 University Of South Florida Inhibitors of akt/pkb with anti-tumor activity
US20090038416A1 (en) 2007-08-07 2009-02-12 Aleta Behrman Bonner System and method for biological sample collection and analyte detection
WO2009032651A1 (en) 2007-08-31 2009-03-12 Smithkline Beecham Corporation Inhibitors of akt activity
WO2009042193A1 (en) 2007-09-25 2009-04-02 Abbott Laboratories Octahydropentalene compounds as chemokine receptor antagonists
US20090163936A1 (en) 2007-12-21 2009-06-25 Chunlin Yang Coated Tissue Engineering Scaffold
US20100042206A1 (en) 2008-03-04 2010-02-18 Icon Medical Corp. Bioabsorbable coatings for medical devices
US8383124B2 (en) * 2008-03-12 2013-02-26 Children's Hospital Medical Center Mobilization of hematopoietic stem cells
EP2300011A4 (en) 2008-05-27 2012-06-20 Dmi Life Sciences Inc METHODS AND THERAPEUTIC COMPOUNDS
WO2010033088A2 (en) * 2008-09-22 2010-03-25 Agency For Science, Technology And Research (A*Star) Method of improving differentiation of chondrogenic progenitor cells
US8314106B2 (en) 2008-12-29 2012-11-20 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
TWI528982B (zh) 2009-03-04 2016-04-11 曼凱公司 改良的乾粉藥物輸送系統
RU2607374C2 (ru) 2009-10-30 2017-01-10 Новозаймс Байофарма Дк А/С Варианты альбумина
CN101914490B (zh) * 2010-08-13 2012-10-10 中国医科大学 一种人羊膜间充质干细胞无血清培养基及其培养方法
MX343160B (es) 2010-09-07 2016-10-26 Ampio Pharmaceuticals Inc Dicetopiperazinas para uso en inhibir hiperpermeabilidad vascular.
US8507496B2 (en) 2010-09-07 2013-08-13 Dmi Acquisition Corp. Treatment of diseases
US9487754B2 (en) * 2010-10-01 2016-11-08 The Trustees Of Columbia University In The City Of New York Derivation of fibrochondrocytes from progenitor cells
WO2012174472A1 (en) 2011-06-17 2012-12-20 Mannkind Corporation High capacity diketopiperazine microparticles
PL2766029T3 (pl) 2011-10-10 2020-08-24 Ampio Pharmaceuticals, Inc. Leczenie choroby zwyrodnieniowej stawów
KR20140075772A (ko) 2011-10-10 2014-06-19 앰피오 파마슈티컬스 인코퍼레이티드 면역 내성을 증가시킨 이식 의료 장비, 및 그의 제조 및 이식 방법
CN103841974A (zh) 2011-10-28 2014-06-04 安皮奥制药股份有限公司 鼻炎的治疗
JP3176478U (ja) 2012-04-11 2012-06-21 ネイス株式会社 運動用エアートランポリン
MX2015009908A (es) 2013-02-01 2015-09-24 Ampio Pharmaceuticals Inc Metodos para producir dicetopiperazinas y composiciones que contienen dicetopiperazinas.
MX2015010937A (es) 2013-03-15 2015-10-29 Ampio Pharmaceuticals Inc Composiciones para la movilizacion, autodireccion, expansion y diferenciacion de celulas madre y metodos para usar las mismas.
CA2958080A1 (en) 2014-08-18 2016-02-25 Ampio Pharmaceuticals, Inc. Treatment of joint conditions
EP3310375A4 (en) 2015-06-22 2019-02-20 Ampio Pharmaceuticals, Inc. USE OF LOW MOLECULAR WEIGHT HUMAN SERUM ALBUMIN FRACTIONS FOR TREATING DISEASES

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4694081A (en) 1985-09-23 1987-09-15 Monsanto Company Process to prepare 2,5-diketopiperazines
WO1996000391A1 (en) 1994-06-23 1996-01-04 Affymax Technologies N.V. Methods for the synthesis of diketopiperazines
US5817751A (en) 1994-06-23 1998-10-06 Affymax Technologies N.V. Method for synthesis of diketopiperazine and diketomorpholine derivatives
WO1997048685A1 (en) 1996-06-18 1997-12-24 Glaxo Group Limited Metalloprotease inhibitors
US5932579A (en) 1996-06-18 1999-08-03 Affymax Technologies N.V. Collagenase-1 and stromelysin-1 inhibitors, pharmaceutical compositions comprising same and methods of their use
US5990112A (en) 1996-06-18 1999-11-23 Affymax Technologies N.V. Inhibitors of metalloproteases pharmaceutical compositions comprising same and methods of their use
US20040024180A1 (en) 2000-04-20 2004-02-05 Karlheinz Drauz Process for the production of 2,5 -diketopiperazines,2,5-diketopiperazines , dipeptides and their use thereof
US6555543B2 (en) 2000-08-04 2003-04-29 Dmi Biosciences, Inc. Method of using diketopiperazines and composition containing them

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"Culture of Hematopoietic Cells", 1994, WILEY-LISS, pages: 81 - 98
BABA ET AL., IMMUNITY, vol. 23, 2005, pages 599 - 609
COLLINS, P. C. ET AL., CURR. OPIN. BIOTECH., vol. 7, 1996, pages 223 - 230
CONG ET AL., J BIOL CHEM, vol. 275, 2000, pages 35665 - 35668
DE FELICE ET AL., CANCER RES, vol. 65, 2005, pages 1505 - 1513
GOTOH ET AL., CELL GROWTH DIFFER, vol. 8, 1997, pages 721 - 729
HE ET AL., CLIN LUNG CANCER, vol. 7, 2005, pages 54 - 60
HOU ET AL., EXP CELL RES, vol. 274, 2002, pages 25 - 34
HOWITZ ET AL., NATURE, vol. 425, 2003, pages 191 - 196
IVANOVIC ET AL., EXP HEMATOL, vol. 30, 2002, pages 67 - 73
LE BLANC ET AL., LANCET, vol. 363, 2004, pages 1439 - 1441
MCADAMS, T. A. ET AL., TIBTECH, vol. 14, 1996, pages 341 - 349
MUKHOPADHYAY ET AL., J CELL MOL MED, vol. 9, 2005, pages 662 - 669
SANDSTROM, E. E. ET AL., BIOTECH. & BIOENGIN., vol. 43, 1994, pages 706 - 733
SATO ET AL., NAT MED, vol. 10, 2004, pages 55 - 63
See also references of EP2968315A4
SMITH ET AL., BIOORG. MED. CHEM. LETTERS, vol. 8, 1998, pages 2369 - 2374

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3183240A4 (en) * 2014-08-18 2018-06-27 Ampio Pharmaceuticals, Inc. Treatment of joint conditions
AU2015305611B2 (en) * 2014-08-18 2020-04-23 Ampio Pharmaceuticals, Inc. Treatment of joint conditions

Also Published As

Publication number Publication date
EP2968315A4 (en) 2016-08-31
EP2968315A2 (en) 2016-01-20
BR112015020469A2 (pt) 2020-01-28
EA201500943A1 (ru) 2016-08-31
CN105101965B (zh) 2021-03-09
SG11201506267XA (en) 2015-09-29
US9808454B2 (en) 2017-11-07
KR20150132508A (ko) 2015-11-25
HK1214143A1 (zh) 2016-07-22
AU2014232728B2 (en) 2019-02-21
AU2014232728A1 (en) 2015-10-29
CN105101965A (zh) 2015-11-25
WO2014145729A3 (en) 2014-12-24
SG10201707619RA (en) 2017-10-30
US20220016110A1 (en) 2022-01-20
JP2016515533A (ja) 2016-05-30
US11026940B2 (en) 2021-06-08
ZA201507291B (en) 2019-04-24
MX2015010937A (es) 2015-10-29
PH12015502048A1 (en) 2016-01-18
EP2968315B1 (en) 2020-06-03
JP6588005B2 (ja) 2019-10-09
US20180153880A1 (en) 2018-06-07
NZ712630A (en) 2021-07-30
CA2906864A1 (en) 2014-09-18
US20140286913A1 (en) 2014-09-25
IL240473A0 (en) 2015-09-24

Similar Documents

Publication Publication Date Title
US20220016110A1 (en) Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
Kalaszczynska et al. Wharton’s jelly derived mesenchymal stem cells: future of regenerative medicine? Recent findings and clinical significance
CN105112357B (zh) 来源于胎盘的贴壁细胞及其在疾病治疗中的应用
US8455245B2 (en) ABCB5 positive mesenchymal stem cells as immunomodulators
EP2118267B1 (en) Novel methods for modulating inflammatory and/or immune responses
Rinaldi et al. Stem cells for skeletal muscle regeneration: therapeutic potential and roadblocks
IL211508A (en) Modulating stem cell and progenitor cell differentiation, vehicle testing and their uses
US10041039B2 (en) Method for producing pluripotent stem cells derived from dental pulp
US20180142211A1 (en) Methods of mesenchymal stem cell mobilization and expansion
Locatelli et al. Mesenchymal stromal cells, from indifferent spectators to principal actors. Are we going to witness a revolution in the scenario of allograft and immune-mediated disorders?
AU2013206755B2 (en) Activating adipose-derived stem cells for transplantation
KR20140071277A (ko) 면역조절 활성을 갖는 세포 집단, 그의 제조 방법 및 용도
WO2016019332A1 (en) Method and apparatus for recovery of umbilical cord tissue derived regenerative cells and uses thereof
JP6410343B2 (ja) 脂肪組織由来幹細胞から表皮角化細胞への誘導
US20160367603A1 (en) Methods for Modulating Skin Inflammation
AU2012200787C1 (en) Modulation of stem and progenitor cell differentiation, assays, and uses thereof
Pappalardo et al. Analysis of Immunomodulatory Properties of Spheroids from Adipose-derived Stem Cells
Pittenger et al. Immunological properties of mesenchymal stem cells isolated from bone marrow and umbilical cord

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201480015832.9

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14763038

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 240473

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/010937

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2016503416

Country of ref document: JP

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015020469

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 12015502048

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2906864

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2014763038

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: IDP00201506507

Country of ref document: ID

WWE Wipo information: entry into national phase

Ref document number: 201500943

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20157029761

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2014232728

Country of ref document: AU

Date of ref document: 20140317

Kind code of ref document: A

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14763038

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 112015020469

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150825

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112015020469

Country of ref document: BR

Kind code of ref document: A2

Free format text: APRESENTE CESSAO DE TODAS AS 5 (CINCO) PRIORIDADES AMERICANAS REIVINDICADAS.

ENP Entry into the national phase

Ref document number: 112015020469

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150825